Overexpression of Eimeria tenella Rhoptry Kinase 2 Induces Early Production of Schizonts

Rhoptries are specialized secretory organelles found in zoite stages of apicomplexan parasites. In addition to well-conserved rhoptry neck proteins, their protein consists mostly of kinase proteins, highly divergent from eukaryotic kinases. ABSTRACT Eimeria tenella is an obligate intracellular parasite responsible for avian coccidiosis. Like other apicomplexan parasites, such as Toxoplasma gondii, cell invasion and intracellular development rely on apical organelle content discharge, named micronemes and rhoptries. Some rhoptry (ROP) kinases (ROPK) are key virulence factors in T. gondii. To date, among the 28 ropk genes carried by E. tenella, only two to four were confirmed by proteomic analysis or immunostaining to be expressed at the sporozoite stage. We have previously shown that EtROP1 is implicated in the inhibition of host cell apoptosis by interacting with the cellular p53. This work functionally described the second ROP kinase expressed at the sporozoite stage in E. tenella. EtROP2 is an active kinase that phosphorylates cell substrates of approximately 50 kDa. Its overexpression leads to the shortening of the prepatent period and to the early development of first-generation schizonts. Conduction of RNA sequencing analysis and reverse transcriptase quantitative PCR (RT-qPCR) on the host cell allowed us to identify the mitogen-activated protein kinase (MAPK) pathway and the transcription factor cFos to be upregulated by EtROP2. We also showed by immunofluorescence assay that the active kinase EtROP2 is implicated in the p38 MAPK pathway activation. We established here that EtROP2 activates the p38 MAPK pathway through a direct or indirect phosphorylation, leading to the overexpression of the master transcription factor cFos known to be implicated in E. tenella development. IMPORTANCE Rhoptries are specialized secretory organelles found in zoite stages of apicomplexan parasites. In addition to well-conserved rhoptry neck proteins, their protein consists mostly of kinase proteins, highly divergent from eukaryotic kinases. Some of those kinases are described as major virulence factors in Toxoplasma gondii, secreted into the host cell to hijack signaling pathways. Most of those kinases remain to be characterized in Eimeria tenella. Deciphering their cellular function is a prerequisite to supporting their relevance as a druggable target in development of new means of Eimeria tenella control. Secreted divergent kinases that interact with host cell partners to modulate pathways are good candidates, as they coevolve with their host targets to ensure their function within the host and are less prone to mutations that would lead to drug resistance. The absence of any orthologous kinase in host cells makes these parasite kinases a promising drug target candidate.

[1]  Xiaomin Zhao,et al.  Identification and Characterization of Eimeria tenella Rhoptry Protein 35 (EtROP35) , 2022, Veterinary sciences.

[2]  L. Hongmei,et al.  Characterization of the Eimeria tenella rhoptry protein with a nuclear localization sequence (EtROP30) , 2022, Parasitology Research.

[3]  Xiaoxin Liu,et al.  Identification and Protective Efficacy of Eimeria tenella Rhoptry Kinase Family Protein 17 , 2022, Animals : an open access journal from MDPI.

[4]  F. Bussière,et al.  Genome-Wide Expression Patterns of Rhoptry Kinases during the Eimeria tenella Life-Cycle , 2021, Microorganisms.

[5]  D. Xia,et al.  Genetic and biological characterisation of three cryptic Eimeria operational taxonomic units that infect chickens (Gallus gallus domesticus)☆ , 2021, International journal for parasitology.

[6]  Namita Singh,et al.  Role of p38 mitogen‐activated protein kinase signalling in virus replication and potential for developing broad spectrum antiviral drugs , 2021, Reviews in medical virology.

[7]  H. Mollenkopf,et al.  Toxoplasma and Eimeria co-opt the host cFos expression for intracellular development in mammalian cells , 2021, Computational and structural biotechnology journal.

[8]  D. Xia,et al.  The Growth of Eimeria tenella: Characterization and Application of Quantitative Methods to Assess Sporozoite Invasion and Endogenous Development in Cell Culture , 2020, Frontiers in Cellular and Infection Microbiology.

[9]  J. Rushton,et al.  Re-calculating the cost of coccidiosis in chickens , 2020, Veterinary Research.

[10]  Qun Liu,et al.  A Novel Rhoptry Protein as Candidate Vaccine against Eimeria tenella Infection , 2020, Vaccines.

[11]  Guang-Hui Zhao,et al.  Genome-wide analysis of differentially expressed profiles of mRNAs, lncRNAs and circRNAs in chickens during Eimeria necatrix infection , 2020, Parasites & Vectors.

[12]  Jared L. Johnson,et al.  Development of a CDK10/CycM in vitro Kinase Screening Assay and Identification of First Small-Molecule Inhibitors , 2020, Frontiers in Chemistry.

[13]  Min Zhang,et al.  Proteomic analysis of the second-generation merozoites of Eimeria tenella under nitromezuril and ethanamizuril stress , 2019, Parasites & vectors.

[14]  M. Roque-Barreira,et al.  Receptor Heterodimerization and Co-Receptor Engagement in TLR2 Activation Induced by MIC1 and MIC4 from Toxoplasma gondii , 2019, International journal of molecular sciences.

[15]  D. Blake,et al.  Laboratory Growth and Genetic Manipulation of Eimeria tenella , 2019, Current protocols in microbiology.

[16]  F. Bussière,et al.  Eimeria tenella ROP kinase EtROP1 induces G0/G1 cell cycle arrest and inhibits host cell apoptosis , 2019, Cellular microbiology.

[17]  F. Bussière,et al.  Establishment of an in vitro chicken epithelial cell line model to investigate Eimeria tenella gamete development , 2018, Parasites & Vectors.

[18]  P. Uhlén,et al.  Voltage-dependent calcium channel signaling mediates GABAA receptor-induced migratory activation of dendritic cells infected by Toxoplasma gondii , 2017, PLoS pathogens.

[19]  J. Dubremetz,et al.  Gliding motility powers invasion and egress in Apicomplexa , 2017, Nature Reviews Microbiology.

[20]  Thomas Cokelaer,et al.  'Sequana': a Set of Snakemake NGS pipelines , 2017, J. Open Source Softw..

[21]  L. Sibley,et al.  Toxoplasma Effectors Targeting Host Signaling and Transcription , 2017, Clinical Microbiology Reviews.

[22]  Jun Wan,et al.  A Human Proteome Array Approach to Identifying Key Host Proteins Targeted by Toxoplasma Kinase ROP18* , 2017, Molecular & Cellular Proteomics.

[23]  M. Ehlers,et al.  Identification of Phosphorylation Consensus Sequences and Endogenous Neuronal Substrates of the Psychiatric Risk Kinase TNIK , 2016, The Journal of Pharmacology and Experimental Therapeutics.

[24]  Matthieu Schapira,et al.  Identification of small molecule inhibitors that block the Toxoplasma gondii rhoptry kinase ROP18. , 2016, ACS infectious diseases.

[25]  Marie-Agnès Dillies,et al.  SARTools: A DESeq2- and EdgeR-Based R Pipeline for Comprehensive Differential Analysis of RNA-Seq Data , 2015, bioRxiv.

[26]  M. Okoniewski,et al.  RNA Seq analysis of the Eimeria tenella gametocyte transcriptome reveals clues about the molecular basis for sexual reproduction and oocyst biogenesis , 2015, BMC Genomics.

[27]  F. Bussière,et al.  Reduced Parasite Motility and Micronemal Protein Secretion by a p38 MAPK Inhibitor Leads to a Severe Impairment of Cell Invasion by the Apicomplexan Parasite Eimeria tenella , 2015, PloS one.

[28]  Tareq B. Malas,et al.  Genomic analysis of the causative agents of coccidiosis in domestic chickens , 2014, Genome research.

[29]  J. Barta,et al.  Coccidiosis: recent advancements in the immunobiology of Eimeria species, preventive measures, and the importance of vaccination as a control tool against these Apicomplexan parasites , 2014, Veterinary medicine.

[30]  H. D. Chapman Milestones in avian coccidiosis research: a review. , 2014, Poultry science.

[31]  A. Bougdour,et al.  A Toxoplasma dense granule protein, GRA24, modulates the early immune response to infection by promoting a direct and sustained host p38 MAPK activation , 2013, The Journal of experimental medicine.

[32]  N. Kannan,et al.  Structural and evolutionary adaptation of rhoptry kinases and pseudokinases, a family of coccidian virulence factors , 2013, BMC Evolutionary Biology.

[33]  Shaohua Zhang,et al.  Transcriptome Analysis in Chicken Cecal Epithelia upon Infection by Eimeria tenella In Vivo , 2013, PloS one.

[34]  A. Pain,et al.  The rhoptry proteome of Eimeria tenella sporozoites. , 2013, International journal for parasitology.

[35]  Natarajan Kannan,et al.  Structural and evolutionary divergence of eukaryotic protein kinases in Apicomplexa , 2011, BMC Evolutionary Biology.

[36]  D. Marc,et al.  A novel chicken lung epithelial cell line: characterization and response to low pathogenicity avian influenza virus. , 2011, Virus Research.

[37]  A. Thomas,et al.  The RON2-AMA1 Interaction is a Critical Step in Moving Junction-Dependent Invasion by Apicomplexan Parasites , 2011, PLoS pathogens.

[38]  D. Roos,et al.  Integrative genomic approaches highlight a family of parasite-specific kinases that regulate host responses. , 2010, Cell host & microbe.

[39]  J. Boothroyd,et al.  Toxoplasma Rhoptry Protein 16 (ROP16) Subverts Host Function by Direct Tyrosine Phosphorylation of STAT6* , 2010, The Journal of Biological Chemistry.

[40]  K. Wimmers,et al.  Microarray-based transcriptional profiling of Eimeria bovis-infected bovine endothelial host cells , 2010, Veterinary research.

[41]  S. Goueli,et al.  ADP-Glo: A Bioluminescent and homogeneous ADP monitoring assay for kinases. , 2009, Assay and drug development technologies.

[42]  Amos Bairoch,et al.  PROSITE, a protein domain database for functional characterization and annotation , 2009, Nucleic Acids Res..

[43]  J. Yates,et al.  Proteomic comparison of four Eimeria tenella life‐cycle stages: Unsporulated oocyst, sporulated oocyst, sporozoite and second‐generation merozoite , 2009, Proteomics.

[44]  E. Phelps,et al.  Toxoplasma gondii Rhoptry Discharge Correlates with Activation of the Early Growth Response 2 Host Cell Transcription Factor , 2008, Infection and Immunity.

[45]  T. Curiel,et al.  Drugs Designed To Inhibit Human p38 Mitogen-Activated Protein Kinase Activation Treat Toxoplasma gondii and Encephalitozoon cuniculi Infection , 2007, Antimicrobial Agents and Chemotherapy.

[46]  G. Labesse,et al.  ROP18 Is a Rhoptry Kinase Controlling the Intracellular Proliferation of Toxoplasma gondii , 2007, PLoS pathogens.

[47]  J. Ajioka,et al.  Polymorphic Secreted Kinases Are Key Virulence Factors in Toxoplasmosis , 2006, Science.

[48]  B. Freedman,et al.  Host cell Ca2+ and protein kinase C regulate innate recognition of Toxoplasma gondii , 2006, Journal of Cell Science.

[49]  B. Freedman,et al.  Host cell Ca2+ and protein kinase C regulate innate recognition of Toxoplasma gondii , 2006, Journal of Cell Science.

[50]  H. Vial,et al.  The rhoptry neck protein RON4 relocalizes at the moving junction during Toxoplasma gondii invasion , 2005, Cellular microbiology.

[51]  S. Brunak,et al.  Improved prediction of signal peptides: SignalP 3.0. , 2004, Journal of molecular biology.

[52]  S. Hanks,et al.  Genomic analysis of the eukaryotic protein kinase superfamily: a perspective , 2003, Genome Biology.

[53]  K. Heidenreich,et al.  Pyridinylimidazole p38 mitogen-activated protein kinase inhibitors block intracellular Toxoplasma gondii replication. , 2002, International Journal of Parasitology.

[54]  L. Sibley,et al.  Cytoskeleton of Apicomplexan Parasites , 2002, Microbiology and Molecular Biology Reviews.

[55]  D. Soldati,et al.  Microneme proteins: structural and functional requirements to promote adhesion and invasion by the apicomplexan parasite Toxoplasma gondii. , 2001, International journal for parasitology.

[56]  N. Mackman,et al.  LPS induction of gene expression in human monocytes. , 2001, Cellular signalling.

[57]  D. Soldati,et al.  Mix and match modules: structure and function of microneme proteins in apicomplexan parasites. , 2001, Trends in parasitology.

[58]  F. Tomley,et al.  Induction of secretion and surface capping of microneme proteins in Eimeria tenella. , 2000, Molecular and biochemical parasitology.

[59]  M. Yaffe,et al.  MAP kinase pathways activated by stress: the p38 MAPK pathway. , 2000, Critical care medicine.

[60]  J. Dubremetz,et al.  Apical organelles and host-cell invasion by Apicomplexa. , 1998, International journal for parasitology.

[61]  D. Fabbro,et al.  Different susceptibility of protein kinases to staurosporine inhibition. Kinetic studies and molecular bases for the resistance of protein kinase CK2. , 1995, European journal of biochemistry.

[62]  A. Bushell,et al.  A live attenuated vaccine for the control of avian coccidiosis: trials in broiler breeders and replacement layer flocks in the United Kingdom , 1995, Veterinary Record.

[63]  T. Hunter,et al.  The eukaryotic protein kinase superfamily: kinase (catalytic) domain structure and classification 1 , 1995, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[64]  M. Karin,et al.  The role of Jun, Fos and the AP-1 complex in cell-proliferation and transformation. , 1991, Biochimica et biophysica acta.

[65]  F. Corpet Multiple sequence alignment with hierarchical clustering. , 1988, Nucleic acids research.

[66]  T. Hunter,et al.  The c-fos protein interacts with c-Jun AP-1 to stimulate transcription of AP-1 responsive genes , 1988, Cell.

[67]  J. Coppee,et al.  SARTools : a DESeq 2-and edgeR-based R pipeline for comprehensive differential analysis of RNA-Seq data , 2015 .

[68]  D. Blake,et al.  Securing poultry production from the ever-present Eimeria challenge. , 2014, Trends in parasitology.

[69]  L. Varuzza,et al.  A comparative transcriptome analysis reveals expression profiles conserved across three Eimeria spp. of domestic fowl and associated with multiple developmental stages. , 2012, International journal for parasitology.

[70]  F. Tomley,et al.  Microneme proteins in apicomplexans. , 2008, Sub-cellular biochemistry.