101
|
Francia ME, Dubremetz JF, Morrissette NS. Basal body structure and composition in the apicomplexans Toxoplasma and Plasmodium. Cilia 2016; 5:3. [PMID: 26855772 PMCID: PMC4743101 DOI: 10.1186/s13630-016-0025-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/20/2016] [Indexed: 11/22/2022] Open
Abstract
The phylum Apicomplexa encompasses numerous important human and animal disease-causing parasites, including the Plasmodium species, and Toxoplasma gondii, causative agents of malaria and toxoplasmosis, respectively. Apicomplexans proliferate by asexual replication and can also undergo sexual recombination. Most life cycle stages of the parasite lack flagella; these structures only appear on male gametes. Although male gametes (microgametes) assemble a typical 9+2 axoneme, the structure of the templating basal body is poorly defined. Moreover, the relationship between asexual stage centrioles and microgamete basal bodies remains unclear. While asexual stages of Plasmodium lack defined centriole structures, the asexual stages of Toxoplasma and closely related coccidian apicomplexans contain centrioles that consist of nine singlet microtubules and a central tubule. There are relatively few ultra-structural images of Toxoplasma microgametes, which only develop in cat intestinal epithelium. Only a subset of these include sections through the basal body: to date, none have unambiguously captured organization of the basal body structure. Moreover, it is unclear whether this basal body is derived from pre-existing asexual stage centrioles or is synthesized de novo. Basal bodies in Plasmodium microgametes are thought to be synthesized de novo, and their assembly remains ill-defined. Apicomplexan genomes harbor genes encoding δ- and ε-tubulin homologs, potentially enabling these parasites to assemble a typical triplet basal body structure. Moreover, the UNIMOD components (SAS6, SAS4/CPAP, and BLD10/CEP135) are conserved in these organisms. However, other widely conserved basal body and flagellar biogenesis elements are missing from apicomplexan genomes. These differences may indicate variations in flagellar biogenesis pathways and in basal body arrangement within the phylum. As apicomplexan basal bodies are distinct from their metazoan counterparts, it may be possible to selectively target parasite structures in order to inhibit microgamete motility which drives generation of genetic diversity in Toxoplasma and transmission for Plasmodium.
Collapse
|
102
|
Abstract
Gametocytes are the specialized form of Plasmodium parasites that are responsible for human-to-mosquito transmission of malaria. Transmission of gametocytes is highly effective, but represents a biomass bottleneck for the parasite that has stimulated interest in strategies targeting the transmission stages separately from those responsible for clinical disease. Studying targets of naturally acquired immunity against transmission-stage parasites may reveal opportunities for novel transmission reducing interventions, particularly the development of a transmission blocking vaccine (TBV). In this review, we summarize the current knowledge on immunity against the transmission stages of Plasmodium. This includes immune responses against epitopes on the gametocyte-infected erythrocyte surface during gametocyte development, as well as epitopes present upon gametocyte activation in the mosquito midgut. We present an analysis of historical data on transmission reducing immunity (TRI), as analysed in mosquito feeding assays, and its correlation with natural recognition of sexual stage specific proteins Pfs48/45 and Pfs230. Although high antibody titres towards either one of these proteins is associated with TRI, the presence of additional, novel targets is anticipated. In conclusion, the identification of novel gametocyte-specific targets of naturally acquired immunity against different gametocyte stages could aid in the development of potential TBV targets and ultimately an effective transmission blocking approach.
Collapse
|
103
|
Santos JM, Kehrer J, Franke-Fayard B, Frischknecht F, Janse CJ, Mair GR. The Plasmodium palmitoyl-S-acyl-transferase DHHC2 is essential for ookinete morphogenesis and malaria transmission. Sci Rep 2015; 5:16034. [PMID: 26526684 PMCID: PMC4630622 DOI: 10.1038/srep16034] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 10/08/2015] [Indexed: 12/20/2022] Open
Abstract
The post-translational addition of C-16 long chain fatty acids to protein cysteine residues is catalysed by palmitoyl-S-acyl-transferases (PAT) and affects the affinity of a modified protein for membranes and therefore its subcellular localisation. In apicomplexan parasites this reversible protein modification regulates numerous biological processes and specifically affects cell motility, and invasion of host cells by Plasmodium falciparum merozoites and Toxoplasma gondii tachyzoites. Using inhibitor studies we show here that palmitoylation is key to transformation of zygotes into ookinetes during initial mosquito infection with P. berghei. We identify DHHC2 as a unique PAT mediating ookinete formation and morphogenesis. Essential for life cycle progression in asexual blood stage parasites and thus refractory to gene deletion analyses, we used promoter swap (ps) methodology to maintain dhhc2 expression in asexual blood stages but down regulate expression in sexual stage parasites and during post-fertilization development of the zygote. The ps mutant showed normal gamete formation, fertilisation and DNA replication to tetraploid cells, but was characterised by a complete block in post-fertilisation development and ookinete formation. Our report highlights the crucial nature of the DHHC2 palmitoyl-S-acyltransferase for transmission of the malaria parasite to the mosquito vector through its essential role for ookinete morphogenesis.
Collapse
Affiliation(s)
- Jorge M Santos
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Edifício Egas Moniz, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Jessica Kehrer
- Parasitology, Department of Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Blandine Franke-Fayard
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Friedrich Frischknecht
- Parasitology, Department of Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Chris J Janse
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Gunnar R Mair
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Edifício Egas Moniz, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal.,Parasitology, Department of Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| |
Collapse
|
104
|
Siqueira-Batista R, Gomes AP, Mendonça EGD, Vitorino RR, Azevedo SFMD, Freitas RDB, Santana LA, Oliveira MGDA. Plasmodium falciparum malaria: proteomic studies. Rev Bras Ter Intensiva 2015; 24:394-400. [PMID: 23917939 PMCID: PMC4031808 DOI: 10.1590/s0103-507x2012000400017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 12/04/2012] [Indexed: 01/21/2023] Open
Abstract
Despite advances in treatment and campaigns for prevention and control of malaria on
the various continents where it is still rampant, this disease remains significantly
relevant to the contemporary world. Plasmodium falciparum is the
organism that is mainly responsible for severe malaria, which is characterized by
disturbances in different organs and systems, with possibly fatal outcomes. Although
incipient, proteomic studies of malaria have yielded favorable prospects for
elucidating the biological aspects of Plasmodium as well as the
pathophysiological, diagnostic, prophylactic, and therapeutic mechanisms of the
disease. Thus, the aim of the present article is to present a brief review of the
applications of proteomic analysis in P. falciparum malaria.
Collapse
|
105
|
Jacot D, Waller RF, Soldati-Favre D, MacPherson DA, MacRae JI. Apicomplexan Energy Metabolism: Carbon Source Promiscuity and the Quiescence Hyperbole. Trends Parasitol 2015; 32:56-70. [PMID: 26472327 DOI: 10.1016/j.pt.2015.09.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 08/28/2015] [Accepted: 09/03/2015] [Indexed: 12/17/2022]
Abstract
The nature of energy metabolism in apicomplexan parasites has been closely investigated in the recent years. Studies in Plasmodium spp. and Toxoplasma gondii in particular have revealed that these parasites are able to employ enzymes in non-traditional ways, while utilizing multiple anaplerotic routes into a canonical tricarboxylic acid (TCA) cycle to satisfy their energy requirements. Importantly, some life stages of these parasites previously considered to be metabolically quiescent are, in fact, active and able to adapt their carbon source utilization to survive. We compare energy metabolism across the life cycle of malaria parasites and consider how this varies in other apicomplexans and related organisms, while discussing how this can be exploited for therapeutic intervention in these diseases.
Collapse
Affiliation(s)
- Damien Jacot
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Ross F Waller
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, UK
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | - James I MacRae
- The Francis Crick Institute, The Ridgeway, Mill Hill, London NW7 1AA, UK.
| |
Collapse
|
106
|
Guttery DS, Roques M, Holder AA, Tewari R. Commit and Transmit: Molecular Players in Plasmodium Sexual Development and Zygote Differentiation. Trends Parasitol 2015; 31:676-685. [PMID: 26440790 DOI: 10.1016/j.pt.2015.08.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 08/04/2015] [Accepted: 08/06/2015] [Indexed: 11/27/2022]
Abstract
During each cycle of asexual endomitotic division in erythrocytes, the malaria parasite makes a fundamental and crucial decision: to continue to invade and proliferate or to differentiate into gametocytes ready for continuation of sexual development. The proteins and regulatory pathways involved in Plasmodium sexual development have been of great interest in recent years as targets for blocking malaria transmission. However, the 'Holy Grail', the master switch orchestrating asexual-to-sexual commitment and further differentiation, has remained elusive - until now. Here we highlight the recent studies identifying the epigenetic and transcriptional master regulators of sexual commitment and discuss the key players in reversible phosphorylation pathways involved in sexual and zygote differentiation.
Collapse
Affiliation(s)
- David S Guttery
- Cell and Developmental Biology Group, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK; Department of Cancer Studies and Cancer Research UK Leicester Centre, University of Leicester, Robert Kilpatrick Building, Leicester Royal Infirmary, Leicester LE2 7LX, UK
| | - Magali Roques
- Cell and Developmental Biology Group, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK
| | - Anthony A Holder
- Mill Hill Laboratory, The Francis Crick Institute, The Ridgeway, Mill Hill, London NW7 1AA, UK
| | - Rita Tewari
- Cell and Developmental Biology Group, School of Life Sciences, Queens Medical Centre, University of Nottingham, Nottingham NG2 7UH, UK.
| |
Collapse
|
107
|
Genome-Wide Collation of the Plasmodium falciparum WDR Protein Superfamily Reveals Malarial Parasite-Specific Features. PLoS One 2015; 10:e0128507. [PMID: 26043001 PMCID: PMC4456382 DOI: 10.1371/journal.pone.0128507] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 04/29/2015] [Indexed: 01/10/2023] Open
Abstract
Despite a significant drop in malaria deaths during the past decade, malaria continues to be one of the biggest health problems around the globe. WD40 repeats (WDRs) containing proteins comprise one of the largest and functionally diverse protein superfamily in eukaryotes, acting as scaffolds for assembling large protein complexes. In the present study, we report an extensive in silico analysis of the WDR gene family in human malaria parasite Plasmodium falciparum. Our genome-wide identification has revealed 80 putative WDR genes in P. falciparum (PfWDRs). Five distinct domain compositions were discovered in Plasmodium as compared to the human host. Notably, 31 PfWDRs were annotated/re-annotated on the basis of their orthologs in other species. Interestingly, most PfWDRs were larger as compared to their human homologs highlighting the presence of parasite-specific insertions. Fifteen PfWDRs appeared specific to the Plasmodium with no assigned orthologs. Expression profiling of PfWDRs revealed a mixture of linear and nonlinear relationships between transcriptome and proteome, and only nine PfWDRs were found to be stage-specific. Homology modeling identified conservation of major binding sites in PfCAF-1 and PfRACK. Protein-protein interaction network analyses suggested that PfWDRs are highly connected proteins with ~1928 potential interactions, supporting their role as hubs in cellular networks. The present study highlights the roles and relevance of the WDR family in P. falciparum, and identifies unique features that lay a foundation for further experimental dissection of PfWDRs.
Collapse
|
108
|
Srivastava A, Creek DJ, Evans KJ, De Souza D, Schofield L, Müller S, Barrett MP, McConville MJ, Waters AP. Host reticulocytes provide metabolic reservoirs that can be exploited by malaria parasites. PLoS Pathog 2015; 11:e1004882. [PMID: 26042734 PMCID: PMC4456406 DOI: 10.1371/journal.ppat.1004882] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 04/14/2015] [Indexed: 11/18/2022] Open
Abstract
Human malaria parasites proliferate in different erythroid cell types during infection. Whilst Plasmodium vivax exhibits a strong preference for immature reticulocytes, the more pathogenic P. falciparum primarily infects mature erythrocytes. In order to assess if these two cell types offer different growth conditions and relate them to parasite preference, we compared the metabolomes of human and rodent reticulocytes with those of their mature erythrocyte counterparts. Reticulocytes were found to have a more complex, enriched metabolic profile than mature erythrocytes and a higher level of metabolic overlap between reticulocyte resident parasite stages and their host cell. This redundancy was assessed by generating a panel of mutants of the rodent malaria parasite P. berghei with defects in intermediary carbon metabolism (ICM) and pyrimidine biosynthesis known to be important for P. falciparum growth and survival in vitro in mature erythrocytes. P. berghei ICM mutants (pbpepc-, phosphoenolpyruvate carboxylase and pbmdh-, malate dehydrogenase) multiplied in reticulocytes and committed to sexual development like wild type parasites. However, P. berghei pyrimidine biosynthesis mutants (pboprt-, orotate phosphoribosyltransferase and pbompdc-, orotidine 5'-monophosphate decarboxylase) were restricted to growth in the youngest forms of reticulocytes and had a severe slow growth phenotype in part resulting from reduced merozoite production. The pbpepc-, pboprt- and pbompdc- mutants retained virulence in mice implying that malaria parasites can partially salvage pyrimidines but failed to complete differentiation to various stages in mosquitoes. These findings suggest that species-specific differences in Plasmodium host cell tropism result in marked differences in the necessity for parasite intrinsic metabolism. These data have implications for drug design when targeting mature erythrocyte or reticulocyte resident parasites.
Collapse
Affiliation(s)
- Anubhav Srivastava
- Wellcome Trust Centre for Molecular Parasitology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, United Kingdom
- Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, United Kingdom
| | - Darren J. Creek
- Wellcome Trust Centre for Molecular Parasitology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, United Kingdom
- Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, United Kingdom
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Krystal J. Evans
- Walter and Eliza Hall Institute of Medical Research, Division of Infection and Immunity, Parkville, Victoria, Australia
| | - David De Souza
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Louis Schofield
- Walter and Eliza Hall Institute of Medical Research, Division of Infection and Immunity, Parkville, Victoria, Australia
- Australian Institute of Tropical Health and Medicine, Centre for Biodiscovery and Molecular Development of Therapeutics, James Cook University, Townsville, Australia
| | - Sylke Müller
- Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, United Kingdom
| | - Michael P. Barrett
- Wellcome Trust Centre for Molecular Parasitology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, United Kingdom
- Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, United Kingdom
| | - Malcolm J. McConville
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Andrew P. Waters
- Wellcome Trust Centre for Molecular Parasitology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, United Kingdom
- Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, United Kingdom
- * E-mail:
| |
Collapse
|
109
|
Reader J, Botha M, Theron A, Lauterbach SB, Rossouw C, Engelbrecht D, Wepener M, Smit A, Leroy D, Mancama D, Coetzer TL, Birkholtz LM. Nowhere to hide: interrogating different metabolic parameters of Plasmodium falciparum gametocytes in a transmission blocking drug discovery pipeline towards malaria elimination. Malar J 2015; 14:213. [PMID: 25994518 PMCID: PMC4449569 DOI: 10.1186/s12936-015-0718-z] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 04/22/2015] [Indexed: 12/23/2022] Open
Abstract
Background The discovery of malaria transmission-blocking compounds is seen as key to malaria elimination strategies and gametocyte-screening platforms are critical filters to identify active molecules. However, unlike asexual parasite assays measuring parasite proliferation, greater variability in end-point readout exists between different gametocytocidal assays. This is compounded by difficulties in routinely producing viable, functional and stage-specific gametocyte populations. Here, a parallel evaluation of four assay platforms on the same gametocyte populations was performed for the first time. This allowed the direct comparison of the ability of different assay platforms to detect compounds with gametocytocidal activity and revealed caveats in some assay readouts that interrogate different parasite biological functions. Methods Gametocytogenesis from Plasmodium falciparum (NF54) was optimized with a robust and standardized protocol. ATP, pLDH, luciferase reporter and PrestoBlue® assays were compared in context of a set of 10 reference compounds. The assays were performed in parallel on the same gametocyte preparation (except for luciferase reporter lines) using the same drug preparations (48 h). The remaining parameters for each assay were all comparable. Results A highly robust method for generating viable and functional gametocytes was developed and comprehensively validated resulting in an average gametocytaemia of 4 %. Subsequent parallel assays for gametocytocidal activity indicated that different assay platforms were not able to screen compounds with variant chemical scaffolds similarly. Luciferase reporter assays revealed that synchronized stage-specific gametocyte production is essential for drug discovery, as differential susceptibility in various gametocyte developmental populations is evident. Conclusions With this study, the key parameters for assays aiming at testing the gametocytocidal activity of potential transmission blocking molecules against Plasmodium gametocytes were accurately dissected. This first and uniquely comparative study emphasizes differential effects seen with the use of different assay platforms interrogating variant biological systems. Whilst this data is informative from a biological perspective and may provide indications of the drug mode of action, it does highlight the care that must be taken when screening broad-diversity chemotypes with a single assay platform against gametocytes for which the biology is not clearly understood. Electronic supplementary material The online version of this article (doi:10.1186/s12936-015-0718-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Janette Reader
- Malaria Parasite Molecular Laboratory, Centre for Sustainable Malaria Control, Department of Biochemistry, University of Pretoria, Private Bag x20, Hatfield, Pretoria, 0028, South Africa.
| | - Mariëtte Botha
- Malaria Parasite Molecular Laboratory, Centre for Sustainable Malaria Control, Department of Biochemistry, University of Pretoria, Private Bag x20, Hatfield, Pretoria, 0028, South Africa.
| | - Anjo Theron
- Biosciences, Council for Scientific and Industrial Research, PO Box 395, Pretoria, 0001, South Africa.
| | - Sonja B Lauterbach
- Plasmodium Molecular Research Unit, Wits Research Institute for Malaria, Department of Molecular Medicine and Haematology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and National Health Laboratory Service, Johannesburg, 2193, South Africa.
| | - Claire Rossouw
- Biosciences, Council for Scientific and Industrial Research, PO Box 395, Pretoria, 0001, South Africa.
| | - Dewaldt Engelbrecht
- Plasmodium Molecular Research Unit, Wits Research Institute for Malaria, Department of Molecular Medicine and Haematology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and National Health Laboratory Service, Johannesburg, 2193, South Africa.
| | - Melanie Wepener
- Plasmodium Molecular Research Unit, Wits Research Institute for Malaria, Department of Molecular Medicine and Haematology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and National Health Laboratory Service, Johannesburg, 2193, South Africa.
| | - Annél Smit
- Malaria Parasite Molecular Laboratory, Centre for Sustainable Malaria Control, Department of Biochemistry, University of Pretoria, Private Bag x20, Hatfield, Pretoria, 0028, South Africa.
| | - Didier Leroy
- Medicines for Malaria Venture, Geneva, Switzerland.
| | - Dalu Mancama
- Biosciences, Council for Scientific and Industrial Research, PO Box 395, Pretoria, 0001, South Africa.
| | - Theresa L Coetzer
- Plasmodium Molecular Research Unit, Wits Research Institute for Malaria, Department of Molecular Medicine and Haematology, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand and National Health Laboratory Service, Johannesburg, 2193, South Africa.
| | - Lyn-Marie Birkholtz
- Malaria Parasite Molecular Laboratory, Centre for Sustainable Malaria Control, Department of Biochemistry, University of Pretoria, Private Bag x20, Hatfield, Pretoria, 0028, South Africa.
| |
Collapse
|
110
|
Alonso-Morales A, González-López L, Cázares-Raga FE, Cortés-Martínez L, Torres-Monzón JA, Gallegos-Pérez JL, Rodríguez MH, James AA, Hernández-Hernández FDLC. Protein phosphorylation during Plasmodium berghei gametogenesis. Exp Parasitol 2015; 156:49-60. [PMID: 26008612 DOI: 10.1016/j.exppara.2015.05.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 05/08/2015] [Accepted: 05/18/2015] [Indexed: 10/23/2022]
Abstract
Plasmodium gametogenesis within the mosquito midgut is a complex differentiation process involving signaling mediated by phosphorylation, which modulate metabolic routes and protein synthesis required to complete this development. However, the mechanisms leading to gametogenesis activation are poorly understood. We analyzed protein phosphorylation during Plasmodium berghei gametogenesis in vitro in serum-free medium using bidimensional electrophoresis (2-DE) combined with immunoblotting (IB) and antibodies specific to phosphorylated serine, threonine and tyrosine. Approximately 75 protein exhibited phosphorylation changes, of which 23 were identified by mass spectrometry. These included components of the cytoskeleton, heat shock proteins, and proteins involved in DNA synthesis and signaling pathways among others. Novel phosphorylation events support a role for these proteins during gametogenesis. The phosphorylation sites of six of the identified proteins, HSP70, WD40 repeat protein msi1, enolase, actin-1 and two isoforms of large subunit of ribonucleoside reductase were investigated using TiO2 phosphopeptides enrichment and tandem mass spectrometry. In addition, transient exposure to hydroxyurea, an inhibitor of ribonucleoside reductase, impaired male gametocytes exflagellation in a dose-dependent manner, and provides a resource for functional studies.
Collapse
Affiliation(s)
- Alberto Alonso-Morales
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Avenida Instituto Politécnico Nacional # 2508, Colonia San Pedro Zacatenco, Delegación Gustavo A. Madero, C.P. 07360, México, D.F., México
| | - Lorena González-López
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Avenida Instituto Politécnico Nacional # 2508, Colonia San Pedro Zacatenco, Delegación Gustavo A. Madero, C.P. 07360, México, D.F., México
| | - Febe Elena Cázares-Raga
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Avenida Instituto Politécnico Nacional # 2508, Colonia San Pedro Zacatenco, Delegación Gustavo A. Madero, C.P. 07360, México, D.F., México
| | - Leticia Cortés-Martínez
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Avenida Instituto Politécnico Nacional # 2508, Colonia San Pedro Zacatenco, Delegación Gustavo A. Madero, C.P. 07360, México, D.F., México
| | - Jorge Aurelio Torres-Monzón
- Centro Regional de Investigación en Salud Pública, Instituto Nacional de Salud Pública, Avenida 19 Poniente esquina 4a Norte s/n, Colonia Centro, C.P. 62100 Tapachula, Chiapas, Mexico
| | | | - Mario Henry Rodríguez
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Avenida Universidad # 655, Colonia Santa María Ahuacatitlán, C.P. 62100, Cuernavaca, Morelos, México
| | - Anthony A James
- Departments of Molecular Biology and Biochemistry, and Microbiology and Molecular Genetics, University of California, Irvine, CA 92697, USA
| | - Fidel de la Cruz Hernández-Hernández
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), Avenida Instituto Politécnico Nacional # 2508, Colonia San Pedro Zacatenco, Delegación Gustavo A. Madero, C.P. 07360, México, D.F., México.
| |
Collapse
|
111
|
Mitochondrial ATP synthase is dispensable in blood-stage Plasmodium berghei rodent malaria but essential in the mosquito phase. Proc Natl Acad Sci U S A 2015; 112:10216-23. [PMID: 25831536 DOI: 10.1073/pnas.1423959112] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Mitochondrial ATP synthase is driven by chemiosmotic oxidation of pyruvate derived from glycolysis. Blood-stage malaria parasites eschew chemiosmosis, instead relying almost solely on glycolysis for their ATP generation, which begs the question of whether mitochondrial ATP synthase is necessary during the blood stage of the parasite life cycle. We knocked out the mitochondrial ATP synthase β subunit gene in the rodent malaria parasite, Plasmodium berghei, ablating the protein that converts ADP to ATP. Disruption of the β subunit gene of the ATP synthase only marginally reduced asexual blood-stage parasite growth but completely blocked mouse-to-mouse transmission via Anopheles stephensi mosquitoes. Parasites lacking the β subunit gene of the ATP synthase generated viable gametes that fuse and form ookinetes but cannot progress beyond this stage. Ookinetes lacking the β subunit gene of the ATP synthase had normal motility but were not viable in the mosquito midgut and never made oocysts or sporozoites, thereby abrogating transmission to naive mice via mosquito bite. We crossed the self-infertile ATP synthase β subunit knockout parasites with a male-deficient, self-infertile strain of P. berghei, which restored fertility and production of oocysts and sporozoites, which demonstrates that mitochondrial ATP synthase is essential for ongoing viability through the female, mitochondrion-carrying line of sexual reproduction in P. berghei malaria. Perturbation of ATP synthase completely blocks transmission to the mosquito vector and could potentially be targeted for disease control.
Collapse
|
112
|
Walker RA, Sharman PA, Miller CM, Lippuner C, Okoniewski M, Eichenberger RM, Ramakrishnan C, Brossier F, Deplazes P, Hehl AB, Smith NC. RNA Seq analysis of the Eimeria tenella gametocyte transcriptome reveals clues about the molecular basis for sexual reproduction and oocyst biogenesis. BMC Genomics 2015; 16:94. [PMID: 25765081 PMCID: PMC4345034 DOI: 10.1186/s12864-015-1298-6] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 01/29/2015] [Indexed: 01/12/2023] Open
Abstract
Background The protozoan Eimeria tenella is a common parasite of chickens, causing avian coccidiosis, a disease of on-going concern to agricultural industries. The high prevalence of E. tenella can be attributed to the resilient oocyst stage, which is transmitted between hosts in the environment. As in related Coccidia, development of the eimerian oocyst appears to be dependent on completion of the parasite’s sexual cycle. RNA Seq transcriptome profiling offers insights into the mechanisms governing the biology of E. tenella sexual stages (gametocytes) and the potential to identify targets for blocking parasite transmission. Results Comparisons between the sequenced transcriptomes of E. tenella gametocytes and two asexual developmental stages, merozoites and sporozoites, revealed upregulated gametocyte transcription of 863 genes. Many of these genes code for proteins involved in coccidian sexual biology, such as oocyst wall biosynthesis and fertilisation, and some of these were characterised in more depth. Thus, macrogametocyte-specific expression and localisation was confirmed for two proteins destined for incorporation into the oocyst wall, as well as for a subtilisin protease and an oxidoreductase. Homologues of an oocyst wall protein and oxidoreductase were found in the related coccidian, Toxoplasma gondii, and shown to be macrogametocyte-specific. In addition, a microgametocyte gamete fusion protein, EtHAP2, was discovered. Conclusions The need for novel vaccine candidates capable of controlling coccidiosis is rising and this panel of gametocyte targets represents an invaluable resource for development of future strategies to interrupt parasite transmission, not just in Eimeria but in other Coccidia, including Toxoplasma, where transmission blocking is a relatively unexplored strategy. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1298-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Robert A Walker
- Queensland Tropical Health Alliance Research Laboratory, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns Campus, McGregor Road, Smithfield, QLD, 4878, Australia. .,Institute of Parasitology, University of Zurich, Winterthurerstrasse 266a, CH-8057, Zürich, Switzerland.
| | - Philippa A Sharman
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Cairns Campus, McGregor Road, Smithfield, QLD, 4878, Australia.
| | - Catherine M Miller
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Cairns Campus, McGregor Road, Smithfield, QLD, 4878, Australia.
| | - Christoph Lippuner
- Institute of Parasitology, University of Zurich, Winterthurerstrasse 266a, CH-8057, Zürich, Switzerland. .,Department of Farm Animal, University of Zurich, Winterthurerstrasse, CH-8057, Zürich, Switzerland.
| | - Michal Okoniewski
- Functional Genomics Center Zurich, Winterthurerstrasse, CH-8057, Zürich, Switzerland.
| | - Ramon M Eichenberger
- Institute of Parasitology, University of Zurich, Winterthurerstrasse 266a, CH-8057, Zürich, Switzerland.
| | - Chandra Ramakrishnan
- Institute of Parasitology, University of Zurich, Winterthurerstrasse 266a, CH-8057, Zürich, Switzerland.
| | - Fabien Brossier
- Apicomplexes et Immunité Mucosale, INRA, UMR1282, Infectiologie et Santé Publique, F-37380, Nouzilly, France. .,Université François Rabelais de Tours, UMR1282, Infectiologie et Santé Publique, F-37000, Tours, France.
| | - Peter Deplazes
- Institute of Parasitology, University of Zurich, Winterthurerstrasse 266a, CH-8057, Zürich, Switzerland.
| | - Adrian B Hehl
- Institute of Parasitology, University of Zurich, Winterthurerstrasse 266a, CH-8057, Zürich, Switzerland.
| | - Nicholas C Smith
- Queensland Tropical Health Alliance Research Laboratory, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns Campus, McGregor Road, Smithfield, QLD, 4878, Australia.
| |
Collapse
|
113
|
Sinden RE. The cell biology of malaria infection of mosquito: advances and opportunities. Cell Microbiol 2015; 17:451-66. [PMID: 25557077 PMCID: PMC4409862 DOI: 10.1111/cmi.12413] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 12/12/2014] [Accepted: 12/24/2014] [Indexed: 01/01/2023]
Abstract
Recent reviews (Feachem et al.; Alonso et al.) have concluded that in order to have a sustainable impact on the global burden of malaria, it is essential that we knowingly reduce the global incidence of infected persons. To achieve this we must reduce the basic reproductive rate of the parasites to < 1 in diverse epidemiological settings. This can be achieved by impacting combinations of the following parameters: the number of mosquitoes relative to the number of persons, the mosquito/human biting rate, the proportion of mosquitoes carrying infectious sporozoites, the daily survival rate of the infectious mosquito and the ability of malaria-infected persons to infect mosquito vectors. This paper focuses on our understanding of parasite biology underpinning the last of these terms: infection of the mosquito. The article attempts to highlight central issues that require further study to assist in the discovery of useful transmission-blocking measures.
Collapse
Affiliation(s)
- R E Sinden
- Department of Life Sciences, Imperial College London and the Jenner Institute, The University of Oxford, Oxford, UK
| |
Collapse
|
114
|
Akinosoglou KA, Bushell ESC, Ukegbu CV, Schlegelmilch T, Cho JS, Redmond S, Sala K, Christophides GK, Vlachou D. Characterization of Plasmodium developmental transcriptomes in Anopheles gambiae midgut reveals novel regulators of malaria transmission. Cell Microbiol 2015; 17:254-68. [PMID: 25225164 PMCID: PMC4371638 DOI: 10.1111/cmi.12363] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 08/27/2014] [Accepted: 09/08/2014] [Indexed: 12/24/2022]
Abstract
The passage through the mosquito is a major bottleneck for malaria parasite populations and a target of interventions aiming to block disease transmission. Here, we used DNA microarrays to profile the developmental transcriptomes of the rodent malaria parasite Plasmodium berghei in vivo, in the midgut of Anopheles gambiae mosquitoes, from parasite stages in the midgut blood bolus to sporulating oocysts on the basal gut wall. Data analysis identified several distinct transcriptional programmes encompassing genes putatively involved in developmental processes or in interactions with the mosquito. At least two of these programmes are associated with the ookinete development that is linked to mosquito midgut invasion and establishment of infection. Targeted disruption by homologous recombination of two of these genes resulted in mutant parasites exhibiting notable infection phenotypes. GAMER encodes a short polypeptide with granular localization in the gametocyte cytoplasm and shows a highly penetrant loss-of-function phenotype manifested as greatly reduced ookinete numbers, linked to impaired male gamete release. HADO encodes a putative magnesium phosphatase with distinctive cortical localization along the concave ookinete periphery. Disruption of HADO compromises ookinete development leading to significant reduction of oocyst numbers. Our data provide important insights into the molecular framework underpinning Plasmodium development in the mosquito and identifies two genes with important functions at initial stages of parasite development in the mosquito midgut.
Collapse
Affiliation(s)
| | | | | | | | - Jee-Sun Cho
- Department of Life Sciences, Imperial College LondonLondon, UK
| | - Seth Redmond
- Department of Life Sciences, Imperial College LondonLondon, UK
| | - Katarzyna Sala
- Department of Life Sciences, Imperial College LondonLondon, UK
| | - George K Christophides
- Department of Life Sciences, Imperial College LondonLondon, UK
- The Cyprus InstituteNicosia, Cyprus
| | - Dina Vlachou
- Department of Life Sciences, Imperial College LondonLondon, UK
- The Cyprus InstituteNicosia, Cyprus
| |
Collapse
|
115
|
Proietti C, Doolan DL. The case for a rational genome-based vaccine against malaria. Front Microbiol 2015; 5:741. [PMID: 25657640 PMCID: PMC4302942 DOI: 10.3389/fmicb.2014.00741] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/06/2014] [Indexed: 12/22/2022] Open
Abstract
Historically, vaccines have been designed to mimic the immunity induced by natural exposure to the target pathogen, but this approach has not been effective for any parasitic pathogen of humans or complex pathogens that cause chronic disease in humans, such as Plasmodium. Despite intense efforts by many laboratories around the world on different aspects of Plasmodium spp. molecular and cell biology, epidemiology and immunology, progress towards the goal of an effective malaria vaccine has been disappointing. The premise of rational vaccine design is to induce the desired immune response against the key pathogen antigens or epitopes targeted by protective immune responses. We advocate that development of an optimally efficacious malaria vaccine will need to improve on nature, and that this can be accomplished by rational vaccine design facilitated by mining genomic, proteomic and transcriptomic datasets in the context of relevant biological function. In our opinion, modern genome-based rational vaccine design offers enormous potential above and beyond that of whole-organism vaccines approaches established over 200 years ago where immunity is likely suboptimal due to the many genetic and immunological host-parasite adaptations evolved to allow the Plasmodium parasite to coexist in the human host, and which are associated with logistic and regulatory hurdles for production and delivery.
Collapse
Affiliation(s)
- Carla Proietti
- Infectious Diseases Program, QIMR Berghofer Medical Research Institute Brisbane, QLD, Australia
| | - Denise L Doolan
- Infectious Diseases Program, QIMR Berghofer Medical Research Institute Brisbane, QLD, Australia
| |
Collapse
|
116
|
Nikolaeva D, Draper SJ, Biswas S. Toward the development of effective transmission-blocking vaccines for malaria. Expert Rev Vaccines 2015; 14:653-80. [PMID: 25597923 DOI: 10.1586/14760584.2015.993383] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The continued global burden of malaria can in part be attributed to a complex lifecycle, with both human hosts and mosquito vectors serving as transmission reservoirs. In preclinical models of vaccine-induced immunity, antibodies to parasite sexual-stage antigens, ingested in the mosquito blood meal, can inhibit parasite survival in the insect midgut as judged by ex vivo functional studies such as the membrane feeding assay. In an era of renewed political momentum for malaria elimination and eradication campaigns, such observations have fueled support for the development and implementation of so-called transmission-blocking vaccines. While leading candidates are being evaluated using a variety of promising vaccine platforms, the field is also beginning to capitalize on global '-omics' data for the rational genome-based selection and unbiased characterization of parasite and mosquito proteins to expand the candidate list. This review covers the progress and prospects of these recent developments.
Collapse
Affiliation(s)
- Daria Nikolaeva
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Headington, Oxford OX3 7DQ, UK
| | | | | |
Collapse
|
117
|
Chia WN, Goh YS, Rénia L. Novel approaches to identify protective malaria vaccine candidates. Front Microbiol 2014; 5:586. [PMID: 25452745 PMCID: PMC4233905 DOI: 10.3389/fmicb.2014.00586] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/17/2014] [Indexed: 12/17/2022] Open
Abstract
Efforts to develop vaccines against malaria have been the focus of substantial research activities for decades. Several categories of candidate vaccines are currently being developed for protection against malaria, based on antigens corresponding to the pre-erythrocytic, blood stage, or sexual stages of the parasite. Long lasting sterile protection from Plasmodium falciparum sporozoite challenge has been observed in human following vaccination with whole parasite formulations, clearly demonstrating that a protective immune response targeting predominantly the pre-erythrocytic stages can develop against malaria. However, most of vaccine candidates currently being investigated, which are mostly subunits vaccines, have not been able to induce substantial (>50%) protection thus far. This is due to the fact that the antigens responsible for protection against the different parasite stages are still yet to be known and relevant correlates of protection have remained elusive. For a vaccine to be developed in a timely manner, novel approaches are required. In this article, we review the novel approaches that have been developed to identify the antigens for the development of an effective malaria vaccine.
Collapse
Affiliation(s)
- Wan Ni Chia
- Singapore Immunology Network, Agency for Science, Technology and Research Singapore, Singapore ; Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore Singapore, Singapore
| | - Yun Shan Goh
- Singapore Immunology Network, Agency for Science, Technology and Research Singapore, Singapore
| | - Laurent Rénia
- Singapore Immunology Network, Agency for Science, Technology and Research Singapore, Singapore ; Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore Singapore, Singapore
| |
Collapse
|
118
|
Guerreiro A, Deligianni E, Santos JM, Silva PAGC, Louis C, Pain A, Janse CJ, Franke-Fayard B, Carret CK, Siden-Kiamos I, Mair GR. Genome-wide RIP-Chip analysis of translational repressor-bound mRNAs in the Plasmodium gametocyte. Genome Biol 2014; 15:493. [PMID: 25418785 PMCID: PMC4234863 DOI: 10.1186/s13059-014-0493-0] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 10/09/2014] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Following fertilization, the early proteomes of metazoans are defined by the translation of stored but repressed transcripts; further embryonic development relies on de novo transcription of the zygotic genome. During sexual development of Plasmodium berghei, a rodent model for human malaria species including P. falciparum, the stability of repressed mRNAs requires the translational repressors DOZI and CITH. When these repressors are absent, Plasmodium zygote development and transmission to the mosquito vector is halted, as hundreds of transcripts become destabilized. However, which mRNAs are direct targets of these RNA binding proteins, and thus subject to translational repression, is unknown. RESULTS We identify the maternal mRNA contribution to post-fertilization development of P. berghei using RNA immunoprecipitation and microarray analysis. We find that 731 mRNAs, approximately 50% of the transcriptome, are associated with DOZI and CITH, allowing zygote development to proceed in the absence of RNA polymerase II transcription. Using GFP-tagging, we validate the repression phenotype of selected genes and identify mRNAs relying on the 5' untranslated region for translational control. Gene deletion reveals a novel protein located in the ookinete crystalloid with an essential function for sporozoite development. CONCLUSIONS Our study details for the first time the P. berghei maternal repressome. This mRNA population provides the developing ookinete with coding potential for key molecules required for life-cycle progression, and that are likely to be critical for the transmission of the malaria parasite from the rodent and the human host to the mosquito vector.
Collapse
Affiliation(s)
- Ana Guerreiro
- />Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Elena Deligianni
- />Institute of Molecular Biology and Biotechnology (IMBB), Foundation of Research and Technology (FORTH), N. Plastira 100, Heraklio, Crete P.C. 71110 Greece
| | - Jorge M Santos
- />Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Patricia AGC Silva
- />Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Christos Louis
- />Institute of Molecular Biology and Biotechnology (IMBB), Foundation of Research and Technology (FORTH), N. Plastira 100, Heraklio, Crete P.C. 71110 Greece
| | - Arnab Pain
- />Pathogen Genomics Laboratory, Computational Bioscience Research Center, King Abdullah University of Science and Technology, Thuwal-Jeddah, Saudi Arabia
| | - Chris J Janse
- />Department of Parasitology, Leiden University Medical Centre, Leiden, The Netherlands
| | | | - Celine K Carret
- />Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Inga Siden-Kiamos
- />Institute of Molecular Biology and Biotechnology (IMBB), Foundation of Research and Technology (FORTH), N. Plastira 100, Heraklio, Crete P.C. 71110 Greece
| | - Gunnar R Mair
- />Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal
- />Parasitology, Department of Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| |
Collapse
|
119
|
Marques SR, Ramakrishnan C, Carzaniga R, Blagborough AM, Delves MJ, Talman AM, Sinden RE. An essential role of the basal body protein SAS-6 in Plasmodium male gamete development and malaria transmission. Cell Microbiol 2014; 17:191-206. [PMID: 25154861 PMCID: PMC4441282 DOI: 10.1111/cmi.12355] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 07/11/2014] [Accepted: 08/19/2014] [Indexed: 01/07/2023]
Abstract
Gametocytes are the sole Plasmodium parasite stages that infect mosquitoes; therefore development of functional gametes is required for malaria transmission. Flagellum assembly of the Plasmodium male gamete differs from that of most other eukaryotes in that it is intracytoplasmic but retains a key conserved feature: axonemes assemble from basal bodies. The centriole/basal body protein SAS-6 normally regulates assembly and duplication of these organelles and its depletion causes severe flagellar/ciliary abnormalities in a diverse array of eukaryotes. Since basal body and flagellum assembly are intimately coupled to male gamete development in Plasmodium, we hypothesized that SAS-6 disruption may cause gametogenesis defects and perturb transmission. We show that Plasmodium berghei sas6 knockouts display severely abnormal male gametogenesis presenting reduced basal body numbers, axonemal assembly defects and abnormal nuclear allocation. The defects in gametogenesis reduce fertilization and render Pbsas6 knockouts less infectious to mosquitoes. Additionally, we show that lack of Pbsas6 blocks transmission from mosquito to vertebrate host, revealing an additional yet undefined role in ookinete to sporulating oocysts transition. These findings underscore the vulnerability of the basal body/SAS-6 to malaria transmission blocking interventions.
Collapse
Affiliation(s)
- Sara R Marques
- Department of Life Sciences, Imperial College of London, London, SW7 2AZ, UK
| | | | | | | | | | | | | |
Collapse
|
120
|
Doolan DL, Apte SH, Proietti C. Genome-based vaccine design: the promise for malaria and other infectious diseases. Int J Parasitol 2014; 44:901-13. [PMID: 25196370 DOI: 10.1016/j.ijpara.2014.07.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 07/30/2014] [Accepted: 07/31/2014] [Indexed: 01/08/2023]
Abstract
Vaccines are one of the most effective interventions to improve public health, however, the generation of highly effective vaccines for many diseases has remained difficult. Three chronic diseases that characterise these difficulties include malaria, tuberculosis and HIV, and they alone account for half of the global infectious disease burden. The whole organism vaccine approach pioneered by Jenner in 1796 and refined by Pasteur in 1857 with the "isolate, inactivate and inject" paradigm has proved highly successful for many viral and bacterial pathogens causing acute disease but has failed with respect to malaria, tuberculosis and HIV as well as many other diseases. A significant advance of the past decade has been the elucidation of the genomes, proteomes and transcriptomes of many pathogens. This information provides the foundation for new 21st Century approaches to identify target antigens for the development of vaccines, drugs and diagnostic tests. Innovative genome-based vaccine strategies have shown potential for a number of challenging pathogens, including malaria. We advocate that genome-based rational vaccine design will overcome the problem of poorly immunogenic, poorly protective vaccines that has plagued vaccine developers for many years.
Collapse
Affiliation(s)
- Denise L Doolan
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia.
| | - Simon H Apte
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Carla Proietti
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| |
Collapse
|
121
|
Tremp AZ, Al-Khattaf FS, Dessens JT. Distinct temporal recruitment of Plasmodium alveolins to the subpellicular network. Parasitol Res 2014; 113:4177-88. [PMID: 25185663 PMCID: PMC4200347 DOI: 10.1007/s00436-014-4093-4] [Citation(s) in RCA: 17] [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: 07/30/2014] [Accepted: 08/25/2014] [Indexed: 11/28/2022]
Abstract
The zoite stages of malaria parasites (merozoite, ookinete and sporozoite) possess a distinctive cortical structure termed the pellicle, which is defined by a double membrane layer named the inner membrane complex (IMC). The IMC is supported by a cytoskeleton of intermediate filaments, termed the subpellicular network (SPN). Plasmodium IMC1 proteins, or alveolins, make up a conserved family of structurally related proteins that comprise building blocks of the SPN. Here, using green fluorescent protein (GFP) tagging in P. berghei, we show that the alveolins PbIMC1c and PbIMC1e are expressed in all three zoite stages. Our data reveal that PbIMC1e is assembled into the SPN concurrent with pellicle development, while PbIMC1c is assembled after pellicle formation. In the sexual stages, these processes are accompanied by different gene expressions from maternal and paternal alleles: PbIMC1e is expressed uniquely from the maternal allele, while PbIMC1c is expressed from the maternal allele in gametocytes, but from both parental alleles during ookinete development. These findings establish biogenesis of the cortical cytoskeleton in Plasmodium to be a complex and dynamic process, involving distinct parental gene expression and chronological recruitment of its protein constituents. While allelic replacement of the pbimc1c and pbimc1e genes with GFP-tagged versions was readily achieved using double crossover homologous recombination, attempts to disrupt these genes by this strategy only resulted in the integration of the selectable marker and GFP reporter into non-specific genomic locations. The recurrent inability to disrupt these genes provides the first genetic evidence that alveolins are necessary for asexual blood-stage parasite development in Plasmodium.
Collapse
Affiliation(s)
- Annie Z. Tremp
- Pathogen Molecular Biology Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT UK
| | - Fatimah S. Al-Khattaf
- Pathogen Molecular Biology Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT UK
- Department of Infection Control, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Johannes T. Dessens
- Pathogen Molecular Biology Department, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT UK
| |
Collapse
|
122
|
SSP3 is a novel Plasmodium yoelii sporozoite surface protein with a role in gliding motility. Infect Immun 2014; 82:4643-53. [PMID: 25156733 DOI: 10.1128/iai.01800-14] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Plasmodium sporozoites develop within oocysts in the mosquito midgut wall and then migrate to the salivary glands. After transmission, they embark on a complex journey to the mammalian liver, where they infect hepatocytes. Proteins on the sporozoite surface likely mediate multiple steps of this journey, yet only a few sporozoite surface proteins have been described. Here, we characterize a novel, conserved sporozoite surface protein (SSP3) in the rodent malaria parasite Plasmodium yoelii. SSP3 is a putative type I transmembrane protein unique to Plasmodium. By using epitope tagging and SSP3-specific antibodies in conjunction with immunofluorescence microscopy, we showed that SSP3 is expressed in mosquito midgut oocyst sporozoites, exhibiting an intracellular localization. In sporozoites derived from the mosquito salivary glands, however, SSP3 localized predominantly to the sporozoite surface as determined by immunoelectron microscopy. However, the ectodomain of SSP3 appeared to be inaccessible to antibodies in nonpermeabilized salivary gland sporozoites. Antibody-induced shedding of the major surface protein circumsporozoite protein (CSP) exposed the SSP3 ectodomain to antibodies in some sporozoites. Targeted deletion of SSP3 adversely affected in vitro sporozoite gliding motility, which, surprisingly, impacted neither their cell traversal capacity, host cell invasion in vitro, nor infectivity in vivo. Together, these data reveal a previously unappreciated complexity of the Plasmodium sporozoite surface proteome and the roles of surface proteins in distinct biological activities of sporozoites.
Collapse
|
123
|
Talman AM, Prieto JH, Marques S, Ubaida-Mohien C, Lawniczak M, Wass MN, Xu T, Frank R, Ecker A, Stanway RS, Krishna S, Sternberg MJE, Christophides GK, Graham DR, Dinglasan RR, Yates JR, Sinden RE. Proteomic analysis of the Plasmodium male gamete reveals the key role for glycolysis in flagellar motility. Malar J 2014; 13:315. [PMID: 25124718 PMCID: PMC4150949 DOI: 10.1186/1475-2875-13-315] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 07/28/2014] [Indexed: 12/22/2022] Open
Abstract
Background Gametogenesis and fertilization play crucial roles in malaria transmission. While male gametes are thought to be amongst the simplest eukaryotic cells and are proven targets of transmission blocking immunity, little is known about their molecular organization. For example, the pathway of energy metabolism that power motility, a feature that facilitates gamete encounter and fertilization, is unknown. Methods Plasmodium berghei microgametes were purified and analysed by whole-cell proteomic analysis for the first time. Data are available via ProteomeXchange with identifier PXD001163. Results 615 proteins were recovered, they included all male gamete proteins described thus far. Amongst them were the 11 enzymes of the glycolytic pathway. The hexose transporter was localized to the gamete plasma membrane and it was shown that microgamete motility can be suppressed effectively by inhibitors of this transporter and of the glycolytic pathway. Conclusions This study describes the first whole-cell proteomic analysis of the malaria male gamete. It identifies glycolysis as the likely exclusive source of energy for flagellar beat, and provides new insights in original features of Plasmodium flagellar organization. Electronic supplementary material The online version of this article (doi:10.1186/1475-2875-13-315) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Arthur M Talman
- Division of Cell and Molecular Biology, Imperial College, London, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Tao D, Ubaida-Mohien C, Mathias DK, King JG, Pastrana-Mena R, Tripathi A, Goldowitz I, Graham DR, Moss E, Marti M, Dinglasan RR. Sex-partitioning of the Plasmodium falciparum stage V gametocyte proteome provides insight into falciparum-specific cell biology. Mol Cell Proteomics 2014; 13:2705-24. [PMID: 25056935 PMCID: PMC4188997 DOI: 10.1074/mcp.m114.040956] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
One of the critical gaps in malaria transmission biology and surveillance is our lack of knowledge about Plasmodium falciparum gametocyte biology, especially sexual dimorphic development and how sex ratios that may influence transmission from the human to the mosquito. Dissecting this process has been hampered by the lack of sex-specific protein markers for the circulating, mature stage V gametocytes. The current evidence suggests a high degree of conservation in gametocyte gene complement across Plasmodium, and therefore presumably for sex-specific genes as well. To better our understanding of gametocyte development and subsequent infectiousness to mosquitoes, we undertook a Systematic Subtractive Bioinformatic analysis (filtering) approach to identify sex-specific P. falciparum NF54 protein markers based on a comparison with the Dd2 strain, which is defective in producing males, and with syntenic male and female proteins from the reanalyzed and updated P. berghei (related rodent malaria parasite) gametocyte proteomes. This produced a short list of 174 male- and 258 female-enriched P. falciparum stage V proteins, some of which appear to be under strong diversifying selection, suggesting ongoing adaptation to mosquito vector species. We generated antibodies against three putative female-specific gametocyte stage V proteins in P. falciparum and confirmed either conserved sex-specificity or the lack of cross-species sex-partitioning. Finally, our study provides not only an additional resource for mass spectrometry-derived evidence for gametocyte proteins but also lays down the foundation for rational screening and development of novel sex-partitioned protein biomarkers and transmission-blocking vaccine candidates.
Collapse
Affiliation(s)
- Dingyin Tao
- From the ‡W. Harry Feinstone Department of Molecular Microbiology and Immunology and the Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, Maryland, USA
| | - Ceereena Ubaida-Mohien
- §Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, Maryland, USA
| | - Derrick K Mathias
- From the ‡W. Harry Feinstone Department of Molecular Microbiology and Immunology and the Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, Maryland, USA
| | - Jonas G King
- From the ‡W. Harry Feinstone Department of Molecular Microbiology and Immunology and the Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, Maryland, USA
| | - Rebecca Pastrana-Mena
- From the ‡W. Harry Feinstone Department of Molecular Microbiology and Immunology and the Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, Maryland, USA
| | - Abhai Tripathi
- From the ‡W. Harry Feinstone Department of Molecular Microbiology and Immunology and the Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, Maryland, USA
| | - Ilana Goldowitz
- ¶Department of Immunology and Infectious Diseases, Harvard School of Public Health, 665 Huntington Avenue, Boston, Massachusetts, USA
| | - David R Graham
- §Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, Maryland, USA
| | - Eli Moss
- ‖The Broad Institute, 7 Cambridge Center, Cambridge, Massachusetts, USA
| | - Matthias Marti
- ¶Department of Immunology and Infectious Diseases, Harvard School of Public Health, 665 Huntington Avenue, Boston, Massachusetts, USA
| | - Rhoel R Dinglasan
- From the ‡W. Harry Feinstone Department of Molecular Microbiology and Immunology and the Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, Maryland, USA;
| |
Collapse
|
125
|
KAF156 is an antimalarial clinical candidate with potential for use in prophylaxis, treatment, and prevention of disease transmission. Antimicrob Agents Chemother 2014; 58:5060-7. [PMID: 24913172 PMCID: PMC4135840 DOI: 10.1128/aac.02727-13] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Renewed global efforts toward malaria eradication have highlighted the need for novel antimalarial agents with activity against multiple stages of the parasite life cycle. We have previously reported the discovery of a novel class of antimalarial compounds in the imidazolopiperazine series that have activity in the prevention and treatment of blood stage infection in a mouse model of malaria. Consistent with the previously reported activity profile of this series, the clinical candidate KAF156 shows blood schizonticidal activity with 50% inhibitory concentrations of 6 to 17.4 nM against P. falciparum drug-sensitive and drug-resistant strains, as well as potent therapeutic activity in a mouse models of malaria with 50, 90, and 99% effective doses of 0.6, 0.9, and 1.4 mg/kg, respectively. When administered prophylactically in a sporozoite challenge mouse model, KAF156 is completely protective as a single oral dose of 10 mg/kg. Finally, KAF156 displays potent Plasmodium transmission blocking activities both in vitro and in vivo. Collectively, our data suggest that KAF156, currently under evaluation in clinical trials, has the potential to treat, prevent, and block the transmission of malaria.
Collapse
|
126
|
Ostareck DH, Naarmann-de Vries IS, Ostareck-Lederer A. DDX6 and its orthologs as modulators of cellular and viral RNA expression. WILEY INTERDISCIPLINARY REVIEWS-RNA 2014; 5:659-78. [PMID: 24788243 DOI: 10.1002/wrna.1237] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 03/19/2014] [Accepted: 03/21/2014] [Indexed: 12/21/2022]
Abstract
DDX6 (Rck/p54), a member of the DEAD-box family of helicases, is highly conserved from unicellular eukaryotes to vertebrates. Functions of DDX6 and its orthologs in dynamic ribonucleoproteins contribute to global and transcript-specific messenger RNA (mRNA) storage, translational repression, and decay during development and differentiation in the germline and somatic cells. Its role in pathways that promote mRNA-specific alternative translation initiation has been shown to be linked to cellular homeostasis, deregulated tissue development, and the control of gene expression in RNA viruses. Recently, DDX6 was found to participate in mRNA regulation mediated by miRNA-mediated silencing. DDX6 and its orthologs have versatile functions in mRNA metabolism, which characterize them as important post-transcriptional regulators of gene expression.
Collapse
Affiliation(s)
- Dirk H Ostareck
- Experimental Research Unit, Department of Intensive Care and Intermediate Care, University Hospital, RWTH Aachen University, Aachen, Germany
| | | | | |
Collapse
|
127
|
Sinha A, Hughes KR, Modrzynska KK, Otto TD, Pfander C, Dickens NJ, Religa AA, Bushell E, Graham AL, Cameron R, Kafsack BF, Williams AE, Llinas M, Berriman M, Billker O, Waters AP. A cascade of DNA-binding proteins for sexual commitment and development in Plasmodium. Nature 2014; 507:253-257. [PMID: 24572359 PMCID: PMC4105895 DOI: 10.1038/nature12970] [Citation(s) in RCA: 307] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 12/19/2013] [Indexed: 12/04/2022]
Abstract
Commitment to and completion of sexual development are essential for malaria parasites (protists of the genus Plasmodium) to be transmitted through mosquitoes. The molecular mechanism(s) responsible for commitment have been hitherto unknown. Here we show that PbAP2-G, a conserved member of the apicomplexan AP2 (ApiAP2) family of DNA-binding proteins, is essential for the commitment of asexually replicating forms to sexual development in Plasmodium berghei, a malaria parasite of rodents. PbAP2-G was identified from mutations in its encoding gene, PBANKA_143750, which account for the loss of sexual development frequently observed in parasites transmitted artificially by blood passage. Systematic gene deletion of conserved ApiAP2 genes in Plasmodium confirmed the role of PbAP2-G and revealed a second ApiAP2 member (PBANKA_103430, here termed PbAP2-G2) that significantly modulates but does not abolish gametocytogenesis, indicating that a cascade of ApiAP2 proteins are involved in commitment to the production and maturation of gametocytes. The data suggest a mechanism of commitment to gametocytogenesis in Plasmodium consistent with a positive feedback loop involving PbAP2-G that could be exploited to prevent the transmission of this pernicious parasite.
Collapse
Affiliation(s)
- Abhinav Sinha
- Wellcome Trust Centre for Molecular Parasitology, University of Glasgow
| | - Katie R. Hughes
- Wellcome Trust Centre for Molecular Parasitology, University of Glasgow
| | | | | | | | | | | | - Ellen Bushell
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Anne L. Graham
- Wellcome Trust Centre for Molecular Parasitology, University of Glasgow
| | - Rachael Cameron
- Wellcome Trust Centre for Molecular Parasitology, University of Glasgow
| | - Bjorn F.C. Kafsack
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - April E. Williams
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, USA
| | - Manuel Llinas
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, USA
| | | | | | - Andrew P. Waters
- Wellcome Trust Centre for Molecular Parasitology, University of Glasgow
| |
Collapse
|
128
|
Andreadaki M, Morgan RN, Deligianni E, Kooij TWA, Santos JM, Spanos L, Matuschewski K, Louis C, Mair GR, Siden-Kiamos I. Genetic crosses and complementation reveal essential functions for the Plasmodium stage-specific actin2 in sporogonic development. Cell Microbiol 2014; 16:751-67. [PMID: 24471657 DOI: 10.1111/cmi.12274] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 12/18/2013] [Accepted: 01/22/2014] [Indexed: 11/30/2022]
Abstract
Malaria parasites have two actin isoforms, ubiquitous actin1 and specialized actin2. Actin2 is essential for late male gametogenesis, prior to egress from the host erythrocyte. Here, we examined whether the two actins fulfil overlapping functions in Plasmodium berghei. Replacement of actin2 with actin1 resulted in partial complementation of the defects in male gametogenesis and, thus, viable ookinetes were formed, able to invade the midgut epithelium and develop into oocysts. However, these remained small and their DNA was undetectable at day 8 after infection. As a consequence sporogony did not occur, resulting in a complete block of parasite transmission. Furthermore, we show that expression of actin2 is tightly controlled in female stages. The actin2 transcript is translationally repressed in female gametocytes, but translated in female gametes. The protein persists until mature ookinetes; this expression is strictly dependent on the maternally derived expression. Genetic crosses revealed that actin2 functions at an early stage of ookinete formation and that parasites lacking actin2 are unable to undergo sporogony in the mosquito midgut. Our results provide insights into the specialized role of actin2 in Plasmodium development in the mosquito and suggest that the two actin isoforms have distinct biological functions.
Collapse
Affiliation(s)
- Maria Andreadaki
- Institute of Molecular Biology and Biotechnology, FORTH, Heraklion, Greece; Department of Biology, University of Crete, Heraklion, Greece
| | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Morahan B, Garcia-Bustos J. Kinase signalling in Plasmodium sexual stages and interventions to stop malaria transmission. Mol Biochem Parasitol 2014; 193:23-32. [PMID: 24509402 DOI: 10.1016/j.molbiopara.2014.01.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 01/22/2014] [Accepted: 01/28/2014] [Indexed: 12/26/2022]
Abstract
The symptoms of malaria, one of the infectious diseases with the highest mortality and morbidity world-wide, are caused by asexual parasites replicating inside red blood cells. Disease transmission, however, is effected by non-replicating cells which have differentiated into male or female gametocytes. These are the forms infectious to mosquito vectors and the insects are the only hosts where parasite sexual reproduction can take place. Malaria is thus a complex infection in which pharmacological treatment of symptoms may still allow transmission for long periods, while pharmacological blockage of infectivity may not cure symptoms. The process of parasite sexual differentiation and development is still being revealed but it is clear that kinase-mediated signalling mechanisms play a significant role. This review attempts to summarise our limited current knowledge on the signalling mechanisms involved in the transition from asexual replication to sexual differentiation and reproduction, with a brief mention to the effects of current treatments on the sexual stages and to some of the difficulties inherent in developing pharmacological interventions to curtail disease transmission.
Collapse
Affiliation(s)
- Belinda Morahan
- Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia.
| | - Jose Garcia-Bustos
- Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
130
|
In vitro development of Haemoproteus parasites: the efficiency of reproductive cells increase during simultaneous sexual process of different lineages. Parasitol Res 2014; 113:1417-23. [DOI: 10.1007/s00436-014-3782-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 01/10/2014] [Indexed: 11/26/2022]
|
131
|
Kramer S. RNA in development: how ribonucleoprotein granules regulate the life cycles of pathogenic protozoa. WILEY INTERDISCIPLINARY REVIEWS-RNA 2013; 5:263-84. [PMID: 24339376 DOI: 10.1002/wrna.1207] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Revised: 10/22/2013] [Accepted: 10/29/2013] [Indexed: 12/11/2022]
Abstract
Ribonucleoprotein (RNP) granules are important posttranscriptional regulators of messenger RNA (mRNA) fate. Several types of RNP granules specifically regulate gene expression during development of multicellular organisms and are commonly referred to as germ granules. The function of germ granules is not entirely understood and probably diverse, but it is generally agreed that one main function is posttranscriptional regulation of gene expression during early development, when transcription is silent. One example is the translational repression of maternally derived mRNAs in oocytes. Here, I hope to show that the need for regulation of gene expression by RNP granules is not restricted to animal development, but plays an equally important role during the development of pathogenic protozoa. Apicomplexa and Trypanosomatidae have complex life cycles with frequent host changes. The need to quickly adapt gene expression to a new environment as well as the ability to suppress translation to survive latencies is critical for successful completion of life cycles. Posttranscriptional gene regulation is not necessarily simpler in protozoa. Apicomplexa surprise with the presence of micro RNA (miRNAs) and upstream open reading frames (µORFs). Trypanosomes have an unusually large repertoire of different RNP granule types. A better understanding of RNP granules in protozoa may help to gain insight into the evolutionary origin of RNP granules: Trypanosomes for example have two types of granules with interesting similarities to animal germ granules.
Collapse
Affiliation(s)
- Susanne Kramer
- Lehrstuhl für Zell- und Entwicklungsbiologie, Biozentrum, Universität Würzburg, Würzburg, Germany
| |
Collapse
|
132
|
Malaria proteomics: insights into the parasite-host interactions in the pathogenic space. J Proteomics 2013; 97:107-25. [PMID: 24140976 DOI: 10.1016/j.jprot.2013.10.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 06/23/2013] [Accepted: 10/08/2013] [Indexed: 11/23/2022]
Abstract
Proteomics is improving malaria research by providing global information on relevant protein sets from the parasite and the host in connection with its cellular structures and specific functions. In the last decade, reports have described biologically significant elements in the proteome of Plasmodium, which are selectively targeted and quantified, allowing for sensitive and high-throughput comparisons. The identification of molecules by which the parasite and the host react during the malaria infection is crucial to the understanding of the underlying pathogenic mechanisms. Hence, proteomics is playing a major role by defining the elements within the pathogenic space between both organisms that change across the parasite life cycle in association with the host transformation and response. Proteomics has identified post-translational modifications in the parasite and the host that are discussed in terms of functional interactions in malaria parasitism. Furthermore, the contribution of proteomics to the investigation of immunogens for potential vaccine candidates is summarized. The malaria-specific technological advances in proteomics are particularly suited now for identifying host-parasite interactions that could lead to promising targets for therapy, diagnosis or prevention. In this review, we examine the knowledge gained on the biology, pathogenesis, immunity and diagnosis of Plasmodium infection from recent proteomic studies. This article is part of a Special Issue entitled: Trends in Microbial Proteomics.
Collapse
|
133
|
Cao Y, Rui B, Wellems DL, Li M, Chen B, Zhang D, Pan W. Identification of piggyBac-mediated insertions in Plasmodium berghei by next generation sequencing. Malar J 2013; 12:287. [PMID: 23961915 PMCID: PMC3765144 DOI: 10.1186/1475-2875-12-287] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 08/18/2013] [Indexed: 01/24/2023] Open
Abstract
Background The piggyBac transposon system provides a powerful forward genetics tool to study gene function in Plasmodium parasites via random insertion mutagenesis and phenotypic screening. The identification of genotype of piggyBac mutants in the Plasmodium genome is thus an indispensable step in forward genetic analysis. Several PCR-based approaches have been used to identify the piggyBac insertion sites in Plasmodium falciparum and Plasmodium berghei, but all are tedious and inefficient. Next generation sequencing can produce large amounts of sequence data and is particularly suitable for genome-wide association studies. In this study, the Next generation sequencing technology was employed to efficiently identify piggyBac insertion sites in the genome of P. berghei. Methods Plasmodium berghei parasites were co-transfected with piggyBac donor and helper plasmids. Initially, the classical inverse PCR method was used to identify the existence of piggyBac insertions in the P. berghei genome. The whole genome of post-transfection parasites was subsequently sequenced with a PCR-free paired-end module using the Illumina HiSeq sequencing system. The two distinct methods (‘BLAST method’ and ‘SOAP method’) were employed to identify piggyBac insertion sites in the P. berghei genome with Illumina sequencing data. All the identified piggyBac insertions were further tested by half-nested PCR. Results The inverse PCR method resulted in a very low yield of ten individual insertions identified. Conversely, 47 piggyBac insertions were identified from about 1 Gb of Illumina sequencing data via the two distinct analysis methods. The majority of identified piggyBac insertions were confirmed by half-nested PCR. In addition, 1,850 single nucleotide polymorphisms were identified through alignment of the Illumina sequencing data of the P. berghei ANKA strain used in this study with the reference genome sequences. Conclusion This study demonstrates that a high-throughput genome sequencing approach is an efficient tool for the identification of piggyBac-mediated insertions in Plasmodium parasites.
Collapse
Affiliation(s)
- Yi Cao
- Department of Tropical Infectious Diseases, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
134
|
Abstract
SUMMARYEimeriais a common genus of apicomplexan parasites that infect diverse vertebrates, most notably poultry, causing serious disease and economic loss. Like all apicomplexans, eimerians have a complex life cycle characterized by asexual divisions that amplify the parasite population in preparation for sexual reproduction. This can be divided into three events: gametocytogenesis, producing gametocytes from merozoites; gametogenesis, producing microgametes and macrogametes from gametocytes; and fertilization of macrogametes by microgametes, producing diploid zygotes with ensuing meiosis completing the sexual phase. Sexual development inEimeriadepends on the differential expression of stage-specific genes, rather than presence or absence of sex chromosomes. Thus, it involves the generation of specific structures and, implicitly, storage of proteins and regulation of protein expression in macrogametes, in preparation for fertilization. InEimeria, the formation of a unique, resilient structure, the oocyst wall, is essential for completion of the sexual phase and parasite transmission. In this review, we piece together the molecular events that underpin sexual reproduction inEimeriaand use additional details from analogous events inPlasmodiumto fill current knowledge gaps. The mechanisms governing sexual stage formation and subsequent fertilization may represent targets for counteracting parasite transmission.
Collapse
|
135
|
Deligianni E, Morgan RN, Bertuccini L, Wirth CC, Silmon de Monerri NC, Spanos L, Blackman MJ, Louis C, Pradel G, Siden-Kiamos I. A perforin-like protein mediates disruption of the erythrocyte membrane during egress of Plasmodium berghei male gametocytes. Cell Microbiol 2013; 15:1438-55. [PMID: 23461714 DOI: 10.1111/cmi.12131] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 01/31/2013] [Accepted: 02/25/2013] [Indexed: 12/31/2022]
Abstract
Successful gametogenesis of the malaria parasite depends on egress of the gametocytes from the erythrocytes within which they developed. Egress entails rupture of both the parasitophorous vacuole membrane and the erythrocyte plasma membrane, and precedes the formation of the motile flagellated male gametes in a process called exflagellation. We show here that egress of the male gametocyte depends on the function of a perforin-like protein, PPLP2. A mutant of Plasmodium berghei lacking PPLP2 displayed abnormal exflagellation; instead of each male gametocyte forming eight flagellated gametes, it produced gametocytes with only one, shared thicker flagellum. Using immunofluorescence and transmission electron microscopy analysis, and phenotype rescue with saponin or a pore-forming toxin, we conclude that rupture of the erythrocyte membrane is blocked in the mutant. The parasitophorous vacuole membrane, on the other hand, is ruptured normally. Some mutant parasites are still able to develop in the mosquito, possibly because the vigorous motility of the flagellated gametes eventually leads to escape from the persisting erythrocyte membrane. This is the first example of a perforin-like protein in Plasmodium parasites having a role in egress from the host cell and the first parasite protein shown to be specifically required for erythrocyte membrane disruption during egress.
Collapse
Affiliation(s)
- Elena Deligianni
- Institute of Molecular Biology and Biotechnology, FORTH, Heraklion, Greece
| | | | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Saeed S, Carter V, Tremp AZ, Dessens JT. Translational repression controls temporal expression of the Plasmodium berghei LCCL protein complex. Mol Biochem Parasitol 2013; 189:38-42. [PMID: 23684590 PMCID: PMC3694310 DOI: 10.1016/j.molbiopara.2013.04.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 04/18/2013] [Accepted: 04/29/2013] [Indexed: 11/06/2022]
Abstract
We have GFP-tagged the LCCL proteins PbLAP4, PbLAP5 and PbLAP6 in Plasmodium berghei. PbLAP4, PbLAP5 and PbLAP6 associate with the crystalloid organelle in ookinetes. Translational repression controls expression of the LCCL protein repertoire in gametocytes.
Plasmodium LCCL proteins comprise a family of six proteins that function as a protein complex and have essential roles in sporozoite transmission. In Plasmodium berghei, family members PbLAP1, PbLAP2 and PbLAP3 have been shown to be expressed in gametocytes and, following gametogenesis and fertilization, to be targeted to distinctive multivesicular organelles termed crystalloids that form in the ookinete. Here, we show by GFP-tagging that PbLAP4, PbLAP5 and PbLAP6, like their family members, are associated with the crystalloids. However, in contrast to their family members, protein expression of PbLAP4, PbLAP5 and PbLAP6 was not detected in gametocytes, even though transcription of the corresponding genes is most prominent in the sexual blood stage parasites. These results suggest that translational repression controls expression of the LCCL protein repertoire and, consequently, the temporal function of the protein complex during P. berghei development in the mosquito.
Collapse
Affiliation(s)
- Sadia Saeed
- Department of Pathogen Molecular Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, United Kingdom
| | | | | | | |
Collapse
|
137
|
Plasmodium cell biology should inform strategies used in the development of antimalarial transmission-blocking drugs. Future Med Chem 2013; 4:2251-63. [PMID: 23234549 DOI: 10.4155/fmc.12.182] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Malaria is a disease with a devastating impact affecting 216 million people each year and causing 655,000 deaths, most of which are children under 5 years old. Recent appreciation that malaria eradication will require novel interventions to target the parasite during transmission from the human host to the mosquito has lead to an exciting surge in activity to develop transmission-blocking drugs and the high-throughput assays to screen for them. This article presents an overview of transmission-stage cell biology and discusses its impact on assay development to provide a context for researchers to evaluate the relative merits/drawbacks of both screening data obtained from current assays and considerations for future assay design. The most recent knowledge of the transmission-blocking properties of current antimalarial classes is also summarized and, underdeveloped targets for transmission-stage drug discovery are highlighted.
Collapse
|
138
|
Abstract
There is an urgent need for the development of new antimalarial drugs with novel modes of actions. The malarial parasite, Plasmodium falciparum, has a relatively small kinome of <100 kinases, with many members exhibiting a high degree of structural divergence from their host counterparts. A number of Plasmodium kinases have recently been shown by reverse genetics to be essential for various parts of the complex parasitic life cycle, and are thus genetically validated as potential targets. Implementation of mass spectrometry-based phosphoproteomics approaches has informed on key phospho-signalling pathways in the parasite. In addition, global phenotypic screens have revealed a large number of putative protein kinase inhibitors with antimalarial potency. Taken together, these investigations point to the Plasmodium kinome as a rich source of potential new targets. In this review, we highlight recent progress made towards this goal.
Collapse
|
139
|
Miao J, Fan Q, Parker D, Li X, Li J, Cui L. Puf mediates translation repression of transmission-blocking vaccine candidates in malaria parasites. PLoS Pathog 2013; 9:e1003268. [PMID: 23637595 PMCID: PMC3630172 DOI: 10.1371/journal.ppat.1003268] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2012] [Accepted: 02/08/2013] [Indexed: 01/01/2023] Open
Abstract
Translational control of gene expression plays an essential role in development. In malaria parasites, translational regulation is critical during the development of specialized transition stages between the vertebrate host and mosquito vector. Here we show that a Pumilio/FBF (Puf) family RNA-binding protein, PfPuf2, is required for the translation repression of a number of transcripts in gametocytes including two genes encoding the transmission-blocking vaccine candidates Pfs25 and Pfs28. Whereas studies to date support a paradigm of Puf-mediated translation regulation through 3' untranslated regions (UTRs) of target mRNAs, this study, for the first time, identifies a functional Puf-binding element (PBE) in the 5'UTR of pfs25. We provide both in vitro and in vivo evidence to demonstrate that PfPuf2 binds to the PBEs in pfs25 and pfs28 to mediate translation repression. This finding provides a renewed view of Pufs as versatile translation regulators and sheds light on their functions in the development of lower branches of eukaryotes.
Collapse
Affiliation(s)
- Jun Miao
- Department of Entomology, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Qi Fan
- Department of Entomology, Pennsylvania State University, University Park, Pennsylvania, United States of America
- Dalian Institute of Biotechnology, Dalian, Liaoning Province, China
| | - Daniel Parker
- Department of Entomology, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Xiaolian Li
- Department of Entomology, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Jianyong Li
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Liwang Cui
- Department of Entomology, Pennsylvania State University, University Park, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
140
|
Ngwa CJ, Scheuermayer M, Mair GR, Kern S, Brügl T, Wirth CC, Aminake MN, Wiesner J, Fischer R, Vilcinskas A, Pradel G. Changes in the transcriptome of the malaria parasite Plasmodium falciparum during the initial phase of transmission from the human to the mosquito. BMC Genomics 2013; 14:256. [PMID: 23586929 PMCID: PMC3640944 DOI: 10.1186/1471-2164-14-256] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Accepted: 04/01/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The transmission of the malaria parasite Plasmodium falciparum from the human to the mosquito is mediated by dormant sexual precursor cells, the gametocytes, which become activated in the mosquito midgut. Because gametocytes are the only parasite stages able to establish an infection in the mosquito, they play a crucial role in spreading the tropical disease. The human-to-mosquito transmission triggers important molecular changes in the gametocytes, which initiate gametogenesis and prepare the parasite for life-cycle progression in the insect vector. RESULTS To better understand gene regulations during the initial phase of malaria parasite transmission, we focused on the transcriptome changes that occur within the first half hour of parasite development in the mosquito. Comparison of mRNA levels of P. falciparum gametocytes before and 30 min following activation using suppression subtractive hybridization (SSH) identified 126 genes, which changed in expression during gametogenesis. Among these, 17.5% had putative functions in signaling, 14.3% were assigned to cell cycle and gene expression, 8.7% were linked to the cytoskeleton or inner membrane complex, 7.9% were involved in proteostasis and 6.4% in metabolism, 12.7% were cell surface-associated proteins, 11.9% were assigned to other functions, and 20.6% represented genes of unknown function. For 40% of the identified genes there has as yet not been any protein evidence.For a subset of 27 genes, transcript changes during gametogenesis were studied in detail by real-time RT-PCR. Of these, 22 genes were expressed in gametocytes, and for 15 genes transcript expression in gametocytes was increased compared to asexual blood stage parasites. Transcript levels of seven genes were particularly high in activated gametocytes, pointing at functions downstream of gametocyte transmission to the mosquito. For selected genes, a regulated expression during gametogenesis was confirmed on the protein level, using quantitative confocal microscopy. CONCLUSIONS The obtained transcriptome data demonstrate the regulations of gene expression immediately following malaria parasite transmission to the mosquito. Our findings support the identification of proteins important for sexual reproduction and further development of the mosquito midgut stages and provide insights into the genetic basis of the rapid adaption of Plasmodium to the insect vector.
Collapse
Affiliation(s)
- Che Julius Ngwa
- Research Center for Infectious Diseases, University of Würzburg, Josef-Schneider-Strasse 2/D15, 97080 Würzburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
141
|
Wierk JK, Langbehn A, Kamper M, Richter S, Burda PC, Heussler VT, Deschermeier C. Plasmodium berghei MAPK1 displays differential and dynamic subcellular localizations during liver stage development. PLoS One 2013; 8:e59755. [PMID: 23544094 PMCID: PMC3609774 DOI: 10.1371/journal.pone.0059755] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Accepted: 02/18/2013] [Indexed: 11/18/2022] Open
Abstract
Mitogen-activated protein kinases (MAPKs) regulate key signaling events in eukaryotic cells. In the genomes of protozoan Plasmodium parasites, the causative agents of malaria, two genes encoding kinases with significant homology to other eukaryotic MAPKs have been identified (mapk1, mapk2). In this work, we show that both genes are transcribed during Plasmodium berghei liver stage development, and analyze expression and subcellular localization of the PbMAPK1 protein in liver stage parasites. Live cell imaging of transgenic parasites expressing GFP-tagged PbMAPK1 revealed a nuclear localization of PbMAPK1 in the early schizont stage mediated by nuclear localization signals in the C-terminal domain. In contrast, a distinct localization of PbMAPK1 in comma/ring-shaped structures in proximity to the parasite's nuclei and the invaginating parasite membrane was observed during the cytomere stage of parasite development as well as in immature blood stage schizonts. The PbMAPK1 localization was found to be independent of integrity of a motif putatively involved in ATP binding, integrity of the putative activation motif and the presence of a predicted coiled-coil domain in the C-terminal domain. Although PbMAPK1 knock out parasites showed normal liver stage development, the kinase may still fulfill a dual function in both schizogony and merogony of liver stage parasites regulated by its dynamic and stage-dependent subcellular localization.
Collapse
Affiliation(s)
- Jannika Katharina Wierk
- Department of Molecular Parasitology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Annette Langbehn
- Department of Molecular Parasitology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Maria Kamper
- Department of Molecular Parasitology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Stefanie Richter
- Department of Molecular Parasitology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | | | - Christina Deschermeier
- Department of Molecular Parasitology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| |
Collapse
|
142
|
Lin JW, Meireles P, Prudêncio M, Engelmann S, Annoura T, Sajid M, Chevalley-Maurel S, Ramesar J, Nahar C, Avramut CMC, Koster AJ, Matuschewski K, Waters AP, Janse CJ, Mair GR, Khan SM. Loss-of-function analyses defines vital and redundant functions of the Plasmodium rhomboid protease family. Mol Microbiol 2013; 88:318-38. [PMID: 23490234 DOI: 10.1111/mmi.12187] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2013] [Indexed: 11/26/2022]
Abstract
Rhomboid-like proteases cleave membrane-anchored proteins within their transmembrane domains. In apicomplexan parasites substrates include molecules that function in parasite motility and host cell invasion. While two Plasmodium rhomboids, ROM1 and ROM4, have been examined, the roles of the remaining six rhomboids during the malaria parasite's life cycle are unknown. We present systematic gene deletion analyses of all eight Plasmodium rhomboid-like proteins as a means to discover stage-specific phenotypes and potential functions in the rodent malaria model, P. berghei. Four rhomboids (ROM4, 6, 7 and 8) are refractory to gene deletion, suggesting an essential role during asexual blood stage development. In contrast ROM1, 3, 9 and 10 were dispensable for blood stage development and exhibited no, subtle or severe defects in mosquito or liver development. Parasites lacking ROM9 and ROM10 showed no major phenotypic defects. Parasites lacking ROM1 presented a delay in blood stage patency following liver infection, but in contrast to a previous study blood stage parasites had similar growth and virulence characteristics as wild type parasites. Parasites lacking ROM3 in mosquitoes readily established oocysts but failed to produce sporozoites. ROM3 is the first apicomplexan rhomboid identified to play a vital role in sporogony.
Collapse
Affiliation(s)
- Jing-Wen Lin
- Leiden Malaria Research Group (Parasitology), Leiden University Medical Centre, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Carvalho TG, Doerig C, Reininger L. Nima- and Aurora-related kinases of malaria parasites. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:1336-45. [PMID: 23462523 DOI: 10.1016/j.bbapap.2013.02.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Accepted: 02/14/2013] [Indexed: 10/27/2022]
Abstract
Completion of the life cycle of malaria parasite requires a succession of developmental stages which vary greatly with respect to proliferation status, implying a tightly regulated control of the parasite's cell cycle, which remains to be understood at the molecular level. Progression of the eukaryotic cell cycle is controlled by members of mitotic kinase of the families CDK (cyclin-dependent kinases), Aurora, Polo and NIMA. Plasmodium parasites possess cyclin-dependent protein kinases and cyclins, which strongly suggests that some of the principles underlying cell cycle control in higher eukaryotes also operate in this organism. However, atypical features of Plasmodium cell cycle organization and important divergences in the composition of the cell cycle machinery suggest the existence of regulatory mechanisms that are at variance with those of higher eukaryotes. This review focuses on several recently described Plasmodium protein kinases related to the NIMA and Aurora kinase families and discusses their functional involvement in parasite's biology. Given their demonstrated essential roles in the erythrocytic asexual cycle and/or sexual stages, these enzymes represent novel potential drug targets for antimalarial intervention aiming at inhibiting parasite replication and/or blocking transmission of the disease. This article is part of a Special Issue entitled: Inhibitors of Protein Kinases (2012).
Collapse
Affiliation(s)
- Teresa Gil Carvalho
- Department of Microbiology, Monash University, Wellington Road, Clayton, VIC 3800, Australia
| | | | | |
Collapse
|
144
|
Lindner SE, Swearingen KE, Harupa A, Vaughan AM, Sinnis P, Moritz RL, Kappe SHI. Total and putative surface proteomics of malaria parasite salivary gland sporozoites. Mol Cell Proteomics 2013; 12:1127-43. [PMID: 23325771 DOI: 10.1074/mcp.m112.024505] [Citation(s) in RCA: 141] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Malaria infections of mammals are initiated by the transmission of Plasmodium salivary gland sporozoites during an Anopheles mosquito vector bite. Sporozoites make their way through the skin and eventually to the liver, where they infect hepatocytes. Blocking this initial stage of infection is a promising malaria vaccine strategy. Therefore, comprehensively elucidating the protein composition of sporozoites will be invaluable in identifying novel targets for blocking infection. Previous efforts to identify the proteins expressed in Plasmodium mosquito stages were hampered by the technical difficulty of separating the parasite from its vector; without effective purifications, the large majority of proteins identified were of vector origin. Here we describe the proteomic profiling of highly purified salivary gland sporozoites from two Plasmodium species: human-infective Plasmodium falciparum and rodent-infective Plasmodium yoelii. The combination of improved sample purification and high mass accuracy mass spectrometry has facilitated the most complete proteome coverage to date for a pre-erythrocytic stage of the parasite. A total of 1991 P. falciparum sporozoite proteins and 1876 P. yoelii sporozoite proteins were identified, with >86% identified with high sequence coverage. The proteomic data were used to confirm the presence of components of three features critical for sporozoite infection of the mammalian host: the sporozoite motility and invasion apparatus (glideosome), sporozoite signaling pathways, and the contents of the apical secretory organelles. Furthermore, chemical labeling and identification of proteins on live sporozoites revealed previously uncharacterized complexity of the putative sporozoite surface-exposed proteome. Taken together, the data constitute the most comprehensive analysis to date of the protein expression of salivary gland sporozoites and reveal novel potential surface-exposed proteins that might be valuable targets for antibody blockage of infection.
Collapse
Affiliation(s)
- Scott E Lindner
- Malaria Program, Seattle Biomedical Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, Washington 98109, USA
| | | | | | | | | | | | | |
Collapse
|
145
|
Talevich E, Tobin AB, Kannan N, Doerig C. An evolutionary perspective on the kinome of malaria parasites. Philos Trans R Soc Lond B Biol Sci 2012; 367:2607-18. [PMID: 22889911 DOI: 10.1098/rstb.2012.0014] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Malaria parasites belong to an ancient lineage that diverged very early from the main branch of eukaryotes. The approximately 90-member plasmodial kinome includes a majority of eukaryotic protein kinases that clearly cluster within the AGC, CMGC, TKL, CaMK and CK1 groups found in yeast, plants and mammals, testifying to the ancient ancestry of these families. However, several hundred millions years of independent evolution, and the specific pressures brought about by first a photosynthetic and then a parasitic lifestyle, led to the emergence of unique features in the plasmodial kinome. These include taxon-restricted kinase families, and unique peculiarities of individual enzymes even when they have homologues in other eukaryotes. Here, we merge essential aspects of all three malaria-related communications that were presented at the Evolution of Protein Phosphorylation meeting, and propose an integrated discussion of the specific features of the parasite's kinome and phosphoproteome.
Collapse
Affiliation(s)
- Eric Talevich
- Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, 120 Green Street, Athens, GA 30602-7229, USA
| | | | | | | |
Collapse
|
146
|
Pasini EM, Braks JA, Fonager J, Klop O, Aime E, Spaccapelo R, Otto TD, Berriman M, Hiss JA, Thomas AW, Mann M, Janse CJ, Kocken CHM, Franke-Fayard B. Proteomic and genetic analyses demonstrate that Plasmodium berghei blood stages export a large and diverse repertoire of proteins. Mol Cell Proteomics 2012. [PMID: 23197789 PMCID: PMC3567864 DOI: 10.1074/mcp.m112.021238] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Malaria parasites actively remodel the infected red blood cell (irbc) by exporting proteins into the host cell cytoplasm. The human parasite Plasmodium falciparum exports particularly large numbers of proteins, including proteins that establish a vesicular network allowing the trafficking of proteins onto the surface of irbcs that are responsible for tissue sequestration. Like P. falciparum, the rodent parasite P. berghei ANKA sequesters via irbc interactions with the host receptor CD36. We have applied proteomic, genomic, and reverse-genetic approaches to identify P. berghei proteins potentially involved in the transport of proteins to the irbc surface. A comparative proteomics analysis of P. berghei non-sequestering and sequestering parasites was used to determine changes in the irbc membrane associated with sequestration. Subsequent tagging experiments identified 13 proteins (Plasmodium export element (PEXEL)-positive as well as PEXEL-negative) that are exported into the irbc cytoplasm and have distinct localization patterns: a dispersed and/or patchy distribution, a punctate vesicle-like pattern in the cytoplasm, or a distinct location at the irbc membrane. Members of the PEXEL-negative BIR and PEXEL-positive Pb-fam-3 show a dispersed localization in the irbc cytoplasm, but not at the irbc surface. Two of the identified exported proteins are transported to the irbc membrane and were named erythrocyte membrane associated proteins. EMAP1 is a member of the PEXEL-negative Pb-fam-1 family, and EMAP2 is a PEXEL-positive protein encoded by a single copy gene; neither protein plays a direct role in sequestration. Our observations clearly indicate that P. berghei traffics a diverse range of proteins to different cellular locations via mechanisms that are analogous to those employed by P. falciparum. This information can be exploited to generate transgenic humanized rodent P. berghei parasites expressing chimeric P. berghei/P. falciparum proteins on the surface of rodent irbc, thereby opening new avenues for in vivo screening adjunct therapies that block sequestration.
Collapse
Affiliation(s)
- Erica M Pasini
- Biomedical Primate Research Centre, 2288 GJ Rijswijk, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Oehring SC, Woodcroft BJ, Moes S, Wetzel J, Dietz O, Pulfer A, Dekiwadia C, Maeser P, Flueck C, Witmer K, Brancucci NMB, Niederwieser I, Jenoe P, Ralph SA, Voss TS. Organellar proteomics reveals hundreds of novel nuclear proteins in the malaria parasite Plasmodium falciparum. Genome Biol 2012. [PMID: 23181666 PMCID: PMC4053738 DOI: 10.1186/gb-2012-13-11-r108] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The post-genomic era of malaria research provided unprecedented insights into the biology of Plasmodium parasites. Due to the large evolutionary distance to model eukaryotes, however, we lack a profound understanding of many processes in Plasmodium biology. One example is the cell nucleus, which controls the parasite genome in a development- and cell cycle-specific manner through mostly unknown mechanisms. To study this important organelle in detail, we conducted an integrative analysis of the P. falciparum nuclear proteome. RESULTS We combined high accuracy mass spectrometry and bioinformatic approaches to present for the first time an experimentally determined core nuclear proteome for P. falciparum. Besides a large number of factors implicated in known nuclear processes, one-third of all detected proteins carry no functional annotation, including many phylum- or genus-specific factors. Importantly, extensive experimental validation using 30 transgenic cell lines confirmed the high specificity of this inventory, and revealed distinct nuclear localization patterns of hitherto uncharacterized proteins. Further, our detailed analysis identified novel protein domains potentially implicated in gene transcription pathways, and sheds important new light on nuclear compartments and processes including regulatory complexes, the nucleolus, nuclear pores, and nuclear import pathways. CONCLUSION Our study provides comprehensive new insight into the biology of the Plasmodium nucleus and will serve as an important platform for dissecting general and parasite-specific nuclear processes in malaria parasites. Moreover, as the first nuclear proteome characterized in any protist organism, it will provide an important resource for studying evolutionary aspects of nuclear biology.
Collapse
|
148
|
Khan SM, Reece SE, Waters AP, Janse CJ, Kaczanowski S. Why are male malaria parasites in such a rush? EVOLUTION MEDICINE AND PUBLIC HEALTH 2012; 2013:3-13. [PMID: 24481180 PMCID: PMC4183958 DOI: 10.1093/emph/eos003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Host immunity selects for the rapid, adaptive, evolution of genes expressed exclusively in male malaria parasites. Analyses of genomic and proteomic data across multiple malaria species reveals rapid adaptive evolution of genes with sex-biased expression in unicellular parasites. Accelerated evolution enables parasites to cope with host immune responses that reduce fertility. Background: Disease-causing organisms are notorious for fast rates of molecular evolution and the ability to adapt rapidly to changes in their ecology. Sex plays a key role in evolution, and recent studies, in humans and other multicellular organisms, document that genes expressed principally or exclusively in males exhibit the fastest rates of adaptive evolution. However, despite the importance of sexual reproduction for many unicellular taxa, sex-biased gene expression and its evolutionary implications have been overlooked. Methods: We analyse genomic data from multiple malaria parasite (Plasmodium) species and proteomic data sets from different parasite life cycle stages. Results: The accelerated evolution of male-biased genes has only been examined in multicellular taxa, but our analyses reveal that accelerated evolution in genes with male-specific expression is also a feature of unicellular organisms. This ‘fast-male’ evolution is adaptive and likely facilitated by the male-biased sex ratio of gametes in the mating pool. Furthermore, we propose that the exceptional rates of evolution we observe are driven by interactions between males and host immune responses. Conclusions: We reveal a novel form of host–parasite coevolution that enables parasites to evade host immune responses that negatively impact upon fertility. The identification of parasite genes with accelerated evolution has important implications for the identification of drug and vaccine targets. Specifically, vaccines targeting males will be more vulnerable to parasite evolution than those targeting females or both sexes.
Collapse
Affiliation(s)
- Shahid M. Khan
- Leiden Malaria Research Group, Department of Parasitology, LUMC, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; Centre for Immunity, Infection and Evolution, Institutes of Evolution, Infection and Immunity, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, UK; Division of Infection and Immunity, Institute of Biomedical Life Sciences & Wellcome Centre for Molecular Parasitology, University of Glasgow, Glasgow G12 8TA, UK; Department of Bioinformatics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106 Warszawa, Poland
| | - Sarah E. Reece
- Leiden Malaria Research Group, Department of Parasitology, LUMC, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; Centre for Immunity, Infection and Evolution, Institutes of Evolution, Infection and Immunity, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, UK; Division of Infection and Immunity, Institute of Biomedical Life Sciences & Wellcome Centre for Molecular Parasitology, University of Glasgow, Glasgow G12 8TA, UK; Department of Bioinformatics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106 Warszawa, Poland
- *Corresponding author. E-mail: ; tel: +44-131-650-5547; fax: +44-131-650-6564
| | - Andrew P. Waters
- Leiden Malaria Research Group, Department of Parasitology, LUMC, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; Centre for Immunity, Infection and Evolution, Institutes of Evolution, Infection and Immunity, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, UK; Division of Infection and Immunity, Institute of Biomedical Life Sciences & Wellcome Centre for Molecular Parasitology, University of Glasgow, Glasgow G12 8TA, UK; Department of Bioinformatics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106 Warszawa, Poland
| | - Chris J. Janse
- Leiden Malaria Research Group, Department of Parasitology, LUMC, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; Centre for Immunity, Infection and Evolution, Institutes of Evolution, Infection and Immunity, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, UK; Division of Infection and Immunity, Institute of Biomedical Life Sciences & Wellcome Centre for Molecular Parasitology, University of Glasgow, Glasgow G12 8TA, UK; Department of Bioinformatics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106 Warszawa, Poland
| | - Szymon Kaczanowski
- Leiden Malaria Research Group, Department of Parasitology, LUMC, Albinusdreef 2, 2333 ZA Leiden, The Netherlands; Centre for Immunity, Infection and Evolution, Institutes of Evolution, Infection and Immunity, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JT, UK; Division of Infection and Immunity, Institute of Biomedical Life Sciences & Wellcome Centre for Molecular Parasitology, University of Glasgow, Glasgow G12 8TA, UK; Department of Bioinformatics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106 Warszawa, Poland
| |
Collapse
|
149
|
Molecular evolution and phylogenetics of rodent malaria parasites. BMC Evol Biol 2012; 12:219. [PMID: 23151308 PMCID: PMC3538709 DOI: 10.1186/1471-2148-12-219] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 10/31/2012] [Indexed: 11/10/2022] Open
Abstract
Background Over the last 6 decades, rodent Plasmodium species have become key model systems for understanding the basic biology of malaria parasites. Cell and molecular parasitology have made much progress in identifying genes underpinning interactions between malaria parasites, hosts, and vectors. However, little attention has been paid to the evolutionary genetics of parasites, which provides context for identifying potential therapeutic targets and for understanding the selective forces shaping parasites in natural populations. Additionally, understanding the relationships between species, subspecies, and strains, is necessary to maximize the utility of rodent malaria parasites as medically important infectious disease models, and for investigating the evolution of host-parasite interactions. Results Here, we collected multi-locus sequence data from 58 rodent malaria genotypes distributed throughout 13 subspecies belonging to P. berghei, P. chabaudi, P. vinckei, and P. yoelii. We employ multi-locus methods to infer the subspecies phylogeny, and use population-genetic approaches to elucidate the selective patterns shaping the evolution of these organisms. Our results reveal a time-line for the evolution of rodent Plasmodium and suggest that all the subspecies are independently evolving lineages (i.e. species). We show that estimates of species-level polymorphism are inflated if subspecies are not explicitly recognized, and detect purifying selection at most loci. Conclusions Our work resolves previous inconsistencies in the phylogeny of rodent malaria parasites, provides estimates of important parameters that relate to the parasite’s natural history and provides a much-needed evolutionary context for understanding diverse biological aspects from the cross-reactivity of immune responses to parasite mating patterns.
Collapse
|
150
|
Sinden RE, Blagborough AM, Churcher T, Ramakrishnan C, Biswas S, Delves MJ. The design and interpretation of laboratory assays measuring mosquito transmission of Plasmodium. Trends Parasitol 2012; 28:457-65. [DOI: 10.1016/j.pt.2012.07.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 07/20/2012] [Accepted: 07/24/2012] [Indexed: 12/14/2022]
|