Apicortin and Its Tentative Role: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Subjects: Microbiology
Contributor:

Apicortin was identified in silico, in 2009, as a characteristic protein of apicomplexans. It combines a partial p25alpha domain with a DCX (doublecortin) one. Based on its occurrence and one of its characteristic domains, it was termed apicortin. Apicortin, when identified, was shown to occur in apicomplexan parasites and in the placozoan animal, Trichoplax adhaerens. The apicomplexan genomes known then contained it without exception. This situation practically has not changed since then; this statement is valid for the newly sequenced genomes and transcriptomes of apicomplexans as well, almost without exception.

  • apicortin
  • Plasmodium
  • Toxoplasma
  • p25alpha domain
  • DCX domain
  • apicomplexa
  • conoid

[1][2][3][4][5][6][7][8][9][10][11][12][13][14][15][16][17][18][19][20][21][22][23][24][25][26][27][28][29][30][31][32]1. Overview

In 2009, apicortin was identified in silico as a characteristic protein of apicomplexans that also occurs in the placozoa, Trichoplax adhaerens. Since then, it has been found that apicortin also occurs in free-living cousins of apicomplexans (chromerids) and in flagellated fungi. It contains a partial p25-α domain and a doublecortin (DCX) domain, both of which have tubulin/microtubule binding properties. Apicortin has been studied experimentally in two very important apicomplexan pathogens, Toxoplasma gondii and Plasmodium falciparum. It is localized in the apical complex in both parasites. In T. gondii, apicortin plays a key role in shaping the structure of a special tubulin polymer, conoid. In both parasites, its absence or downregulation has been shown to impair pathogen–host interactions. Based on these facts, it has been suggested as a therapeutic target for treatment of malaria and toxoplasmosis.

2. Name

Apicortin was identified in silico, in 2009, as a characteristic protein of apicomplexans [1]. It combines a partial p25alpha domain with a DCX (doublecortin) one. Based on its occurrence and one of its characteristic domains, it was termed apicortin.

3. Apicortin

Apicortin, when identified, was shown to occur in apicomplexan parasites and in the placozoan animal, Trichoplax adhaerens [1]. The apicomplexan genomes known then contained it without exception. This situation practically has not changed since then; this statement is valid for the newly sequenced genomes and transcriptomes of apicomplexans as well. The only exception is the Apicomplexa with the smallest genome, Babesia microti [2].
Later it has been found that apicortin also occurs in chromerids, the recently discovered [3,4], free-living cousins of apicomplexans [5]. This is not surprising, given the phylogenetic proximity and the structural similarity of these phyla. Unlike apicomplexan species, both Chromera velia and Vitrella brassicaformis have three apicortin paralogs. Apicortin has not been found in other related phyla of the Alveolata superphylum, although its remnant is present in the genome in the case of Perkinsozoa [6]. However, the very recently published draft genome of Perkinsus olseni contains hypothetical protein(s) possessing both p25alpha and DCX domains (KAF4710163, KAF4750811) [7].
It has also been revealed that some primitive fungi also possess this protein; first it was shown in the cases of Spizellomyces punctatus [6] and Rozella allomycis [5]. Later, a systematic examination of fungal genomes showed that the flagellated fungi contain apicortin almost without exception; and it is present even in a non-flagellated but also deeper branching clade (Endogonomycetes) [8].
Apicortin is one of the most abundant proteins of T. adhaerens [9]. This is the only animal that possesses apicortin [10]. Animal draft genomes and transcriptomes contain sometimes nucleotide sequences, contigs and TSAs (transcriptome shot-gun assemblies), homologous to apicortin, but they have been shown to be contaminations from parasitizing apicomplexans, based on sequence similarities and GC ratios [11,12]. In the case of T. adhaerens, this option was excluded by phylogenetic analysis [8]. Several authors suggested that the presence of frequent contaminants could serve as a basis for the identification of hitherto unknown apicomplexans and, in general, parasites [13,14,15]. The origin of a few plant and algal apicortin-like nucleotide sequences needs further investigations. My preliminary data suggest that contamination of apicomplexan origin can be ruled out but not that of fungal ones.
Strong correlation between the presence of the p25alpha domain and that of the eukaryotic flagellum was suggested before the identification of apicortin [16]. With very few exceptions, each flagellated organism contains p25alpha domain-containing proteins; on the other side, in non-flagellated species, the p25alpha domain generally does not occur [8,10]. The protein that contains the p25alpha domain varies depending on the phylum; e.g., it is the so called “long-type” TPPP in animals (except T. adhaerens) [10,17], a fungal-type TPPP and apicortin in flagellated fungi [8], while the “short-type” TPPP and apicortin are found in apicomplexan species [10]. With the exceptions of two non-flagellated fungi and some apicomplexans, apicortin can be found only in species which are flagellated, at least in some life stage.

4. Domains

Apicortin belongs to a eukaryotic protein superfamily, the TPPP-like proteins, characterized by the presence of the p25alpha domain (Pfam05517, IPR008907), and named after the first identified member, TPPP/p25, which exhibits microtubule stabilizing function [10]. TPPP stands for tubulin polymerization promoting protein [18,19] and was first identified by Takahashi et al. as p25 protein [20]. Full-length p25alpha domain consists of about 160 amino acids; however, that of the apicortin is incomplete, containing only the last 30–40 amino acids (a “partial p25alpha domain”). Importantly, the tubulin/microtubule binding amino acid sequence is located in this part of the domain [21,22,23,24]. This is the most conserved part of the domain, which contains a characteristic “Rossmann-like motif”, GXGXGXXGR [10,17].
However, apicortins possess another characteristic domain, the DCX one (Pfam03607, IPR003533) [1]. The DCX (doublecortin) domain is named after the brain-specific X-linked gene doublecortin [25]. It is a structural domain, which generally appears in duplicate as two tandemly repeated 80 amino acid regions (N- and C-terminal type DCX domains), but proteins containing only one DCX-repeat have also been identified [26,27]. This domain is also known to play a role in the stabilization of microtubules [25,26]. 

5. Conclusions

When apicortin was identified in silico, it was proposed that its function may be to stabilize specific cytoskeletal structures that are unique to apicomplexans [1]. It has also been hypothesized that it might have a role in parasite–host interactions [1,5]. Recent experimental results support both assumptions [28,29,30,31]. Apicortin has been shown to be localized in the apical complex; more precisely, in T. gondii, it is localized exclusively at the conoid, a tubulin-based structure that has an important role in host cell invasion [29]. Apicortin is essential for providing the correct structure and function of conoid. In P. falciparum, which has no conoid, apicortin is localized at the apical end; it has been suggested that it is involved in the formation of the apical complex [30]. In both species, downregulation of apicortin leads to impaired host cell invasion [28,31].
Apicomplexan species possess several hundred genes, which are specific for the phylum, and even more genes, which are absent in mammals/vertebrates but are present in these parasites. For example, apicomplexans have evolved hundreds of specialized invasion proteins, and contain lineage- and species-specific gene families, which are specialized for modulating host-specific adaptation [32]. Targeting parasite proteins that have crucial roles in these interactions is a key focus in the development of therapeutic agents against diseases caused by apicomplexan infection. Thus, apicortin, which seems to influence parasite–host interactions, is also a potential drug target. Since some mechanistic details of its function have been known, this protein has become more than a desirable target in drug development.

This entry is adapted from the peer-reviewed paper 10.3390/tropicalmed6030118

References

  1. Orosz, F. Apicortin, a unique protein, with a putative cytoskeletal role, shared only by apicomplexan parasites and the plac-ozoan Trichoplax adhaerens. Infect. Genet. Evol. 2009, 9, 1275–1286, doi:10.1016/j.meegid.2009.09.001.
  2. Cornillot, E.; Hadj-Kaddour, K.; Dassouli, A.; Noel, B.; Ranwez, V.; Vacherie, B.; Augagneur, Y.; Brès, V.; Duclos, A.; Randazzo, S.; et al. Sequencing of the smallest Apicomplexan genome from the human pathogen Babesia microti. Nucleic Acids Res. 2012, 40, 9102–9114, doi:10.1093/nar/gks700.
  3. Moore, R.B.; Oborník, M.; Janouškovec, J.; Chrudimský, T.; Vancová, M.; Green, D.H.; Wright, S.W.; Davies, N.W.; Bolch, C.J.; Heimann, K.; et al. A photosynthetic alveolate closely related to apicomplexan parasites. Nature 2008, 451, 959–963, doi:10.1038/nature06635.
  4. Oborník, M.; Modrý, D.; Lukeš, M.; Cernotíková-Stříbrná, E.; Cihlář, J.; Tesařová, M.; Kotabová, E.; Vancová, M.; Prášil, O.; Lukeš, J. Morphology ultrastructure and life cycle of Vitrella brassicaformis n. sp., n. gen., a novel chromerid from the Great Barrier Reef. Protist 2012, 163, 306–323, doi:10.1016/j.protis.2011.09.001.
  5. Orosz, F. Apicomplexan apicortins possess a long disordered N-terminal extension. Infect. Genet. Evol. 2011, 11, 1037–1044, doi:10.1016/j.meegid.2011.03.023.
  6. Bogema, D.R.; Yam, J.; Micallef, M.L.; Gholipourkanani, H.; Go, J.; Jenkins, C.; Dang, C. Draft genomes of Perkinsus olseni and Perkinsus chesapeaki reveal polyploidy and regional differences in heterozygosity. Genomics 2021, 113, 677–688, doi:10.1016/j.ygeno.2020.09.064.
  7. Orosz, F. Wider than thought phylogenetic occurrence of apicortin, a characteristic protein of apicomplexan parasites. J. Mol. Evol. 2016, 82, 303–314, doi:10.1007/s00239-016-9749-5.
  8. Orosz, F. On the TPPP-like proteins of flagellated fungi. Fungal Biol. 2021, 125, 357–367, doi:10.1016/j.funbio.2020.12.001.
  9. Ringrose, J.H.; van den Toorn, H.W.P.; Eitel, M.; Post, H.; Neerincx, P.; Schierwater, B.; Altelaar, A.F.M.; Heck, A.J.R. Deep proteome profiling of Trichoplax adhaerens reveals remarkable features at the origin of metazoan multicellularity. Nat. Commun. 2013, 4, 1408, doi:10.1038/ncomms2424.
  10. Orosz, F. A new protein superfamily: TPPP-like proteins. PLoS ONE 2012, 7, e49276, doi:10.1371/journal.pone.0049276.
  11. Orosz, F. Two recently sequenced vertebrate genomes are contaminated with apicomplexan species of the Sarcocystidae family. Int. J. Parasitol. 2015, 45, 871–878, doi:10.1016/j.ijpara.2015.07.002.
  12. Orosz, F. Does apicortin, a characteristic protein of apicomplexan parasites and placozoa, occur in Eumetazoa? Acta Parasitol. 2018, 63, 617–633, doi:10.1515/ap-2018-0071.
  13. Borner, J.; Burmester, T. Parasite infection of public databases: A data mining approach to identify apicomplexan contamina-tions in animal genome and transcriptome assemblies. BMC Genomics 2017, 18, 100, doi:10.1186/s12864-017-3504-1.
  14. Lopes, R.J.; Mérida, A.M.; Carneiro, M. Unleashing the potential of public genomic resources to find parasite genetic data. Trends Parasitol. 2017, 33, 750–753, doi:10.1016/j.pt.2017.06.006.
  15. Orosz, F. On the benefit of publishing uncurated genome assembly data. J. Bacteriol. Parasitol. 2017, 8, 317, doi:10.4172/2155-9597.1000317.
  16. Orosz, F.; Ovádi, J. TPPP orthologs are ciliary proteins. FEBS Lett. 2008, 582, 3757–3764, doi:10.1016/j.febslet.2008.10.011.
  17. Vincze, O.; Tőkési, N.; Oláh, J.; Hlavanda, E.; Zotter, Á.; Horváth, I.; Lehotzky, A.; Tirián, L.; Medzihradszky, K.F.; Kovács, J.; et al. Tubulin polymerization promoting proteins (TPPPs): Members of a new family with distinct structures and functions. Biochemistry 2006, 45, 13818–13826, doi:10.1021/bi061305e.
  18. Hlavanda, E.; Kovács, J.; Oláh, J.; Orosz, F.; Medzihradszky, K.F.; Ovádi, J. Brain-specific p25 protein binds to tubulin and microtubules and induces aberrant microtubule assemblies at substoichiometric concentrations. Biochemistry 2002, 41, 8657–8664, doi:10.1021/bi020140g.
  19. Tirián, L.; Hlavanda, E.; Oláh, J.; Horváth, I.; Orosz, F.; Szabó, B.; Kovács, J.; Szabad, J.; Ovádi, J. TPPP/p25 promotes tubulin assemblies and blocks mitotic spindle formation. Proc. Natl. Acad. Sci. USA 2003, 100, 13976–13981, doi:10.1073/pnas.2436331100.
  20. Takahashi, M.; Tomizawa, K.; Ishiguro, K.; Sato, K.; Omori, A.; Sato, S.; Shiratsuchi, A.; Uchida, T.; Imahori, K. A novel brainspecific 25 kDa protein (p25) is phosphorylated by a Ser/Thr-Pro kinase (TPK II) from tau protein kinase fractions. FEBS Lett. 1991, 289, 37–43, doi:10.1016/0014-5793(91)80903-g.
  21. Hlavanda, E.; Klement, E.; Kókai, E.; Kovács, J.; Vincze, O.; Tőkési, N.; Orosz, F.; Medzihradszky, K.F.; Dombrádi, V.; Ovádi, J. Phosphorylation blocks the activity of tubulin polymerization-promoting protein TPPP: Identification of sites targeted by dif-ferent kinases. J. Biol. Chem. 2007, 282, 29531–29539, doi:10.1074/jbc.M703466200.
  22. Tőkési, N.; Oláh, J.; Hlavanda, E.; Szunyogh, S.; Szabó, A.; Babos, F.; Magyar, A.; Lehotzky, A.; Vass, E.; Ovádi, J. Identification of motives mediating alternative functions of the neomorphic moonlighting TPPP/p25. Biochim. Biophys. Acta 2014, 1842, 547–557, doi:10.1016/j.bbadis.2014.01.009.
  23. DeBonis, S.; Neumann, E.; Skoufias, D. Self protein-protein interactions are involved in TPPP/p25 mediated microtubule bundling. Sci. Rep. 2015, 5, 13242, doi:10.1038/srep13242.
  24. Oláh, J.; Szénási, T.; Szabó, A.; Kovács, K.; Lőw, P.; Štifanić, M.; Orosz, F. Tubulin binding and polymerization promoting properties of TPPP proteins are evolutionarily conserved. Biochemistry 2017, 56, 1017–1024, doi:10.1021/acs.biochem.6b00902.
  25. Sapir, T.; Horesh, D.; Caspi, M.; Atlas, R.; Burgess, H.A. Doublecortin mutations cluster in evolutionarily conserved functional domains. Hum. Mol. Genet. 2000, 9, 703–712, doi:10.1093/hmg/9.5.703.
  26. Kim, M.H.; Cierpicki, T.; Derewenda, U.; Krowarsch, D.; Feng, Y.; Devedjiev, Y.; Dauter, Z.; Walsh, C.A.; Otlewski, J.; Bushweller, J.H.; et al. The DCX-domain tandems of doublecortin and doublecortin-like kinase. Nat. Struct. Biol. 2003, 10, 324–333, doi:10.1038/nsb918.
  27. Reiner, O.; Coquelle, F.M.; Peter, B.; Levy, T.; Kaplan, A.; Sapir, T.; Orr, I.; Barkai, N.; Eichele, G.; Bergmann, S. The evolving doublecortin (DCX) superfamily. BMC Genome 2006, 7, 188, doi:10.1186/1471-2164-7-188.
  28. Leung, J.M.; Nagayasu, E.; Hwang, Y.C.; Liu, J.; Pierce, P.G.; Phan, I.Q.; Prentice, R.A.; Murray, J.M.; Hu, K. A double-cortin-domain protein of Toxoplasma and its orthologues bind to and modify the structure and organization of tubulin polymers. BMC Mol. Cell Biol. 2020, 21, 8, doi:10.1186/s12860-020-0249-5.
  29. Nagayasu, E.; Hwang, Y.C.; Liu, J.; Murray, J.M.; Hu, K. Loss of a doublecortin (DCX)-domain protein causes structural defects in a tubulin-based organelle of Toxoplasma gondii and impairs host-cell invasion. Mol. Biol. Cell 2017, 28, 411–428, doi:10.1091/mbc.E16-08-0587.
  30. Chakrabarti, M.; Joshi, M.; Kumari, G.; Singh, P.; Shoaib, R.; Munjal, A.; Kumar, V.; Behl, A.; Abid, M.; Garg, S.; et al. Inter-action of Plasmodium falciparum apicortin with α- and β-tubulin is critical for parasite growth and survival. Sci. Rep. 2021, 11, 4688, doi:10.1038/s41598-021-83513-5.
  31. Chakrabarti, M.; Garg, S.; Rajagopal, A.; Pati, S.; Singh, S. Targeted repression of Plasmodium apicortin by host microRNA impairs malaria parasite growth and invasion. Dis. Models Mech. 2020, 13, dmm042820, doi:10.1242/dmm.042820.
  32. Swapna, L.S.; Parkinson, J. Genomics of apicomplexan parasites. Crit. Rev. Biochem. Mol. Biol. 2017, 52, 254–273, doi:10.1080/10409238.2017.1290043.
More
This entry is offline, you can click here to edit this entry!