INTRODUCTION
Infections with protozoan parasites of the genera Plasmodium, Leishmania, Toxoplasma, and Trypanosoma are among the most prevalent diseases in developing countries. Transmission of Plasmodia to human hosts through the bites of infected female Anopheles mosquitoes results in the acute febrile illness malaria. In 2015, 95 countries reported ongoing transmissions, resulting in half of the world's population (3·2 billion people) being at risk of malaria. Plasmodium falciparum and Plasmodium vivax pose the greatest threats with P. falciparum being responsible for most malaria-related deaths and P. vivax being the most dominant malaria parasite outside of sub-Saharan Africa (World Health Organization, 2015). Protozoan parasites of the genus Leishmania, transmitted by the female sand fly, cause the spectrum of diseases known as the leishmaniases. Symptoms range from skin ulcers with permanent scars to the swelling of the spleen and liver. Leishmaniases have been reported in Asia, Africa, South and Central America, and southern Europe, with 20–30 000 deaths annually (World Health Organization, 2016a ). Toxoplasmosis results from an infection with Toxoplasma gondii, transmitted through poorly cooked food, excrements from infected animals, or during pregnancy. Although up to half of the world's population becomes infected at some point in their lives, the immune system can usually cope with the parasite (Flegr et al. Reference Flegr, Prandota, Sovičková and Israili2014). However, toxoplasmosis can cause miscarriage during pregnancy or serious infections of the lungs or brain in people with a weak immune system (Jones et al. Reference Jones, Parise and Fiore2014). Another well-known disease caused by a protozoan parasite is human African trypanosomiasis (HAT) which is caused by Trypanosoma brucei. Also known as sleeping sickness, HAT is transmitted by the tsetse fly and occurs in 36 sub-Saharan African countries (World Health Organization, 2016b ). These diseases have serious and sometimes lethal consequences if untreated. One major challenge is the increasing number of strains that have developed resistance against frontline therapeutics, including chloroquine, pyrimethamine/sulfadoxine, and artemisinin in the case of malaria (Sinha et al. Reference Sinha, Medhi and Sehgal2014; Mbengue et al. Reference Mbengue, Bhattacharjee, Pandharkar, Liu, Estiu, Stahelin, Rizk, Njimoh, Ryan, Chotivanich, Nguon, Ghorbal, Lopez-Rubio, Pfrender, Emrich, Mohandas, Dondorp, Wiest and Haldar2015), pentavalent antimonials in the case of Leishmaniasis (Hajjaran et al. Reference Hajjaran, Kazemi-Rad, Mohebali, Oshaghi, Khadem-Erfan, Hajaliloo, Reisi Nafchi and Raoofian2016), and melarsoprol and pentamidine in the case of HAT (Baker et al. Reference Baker, de Koning, Mäser and Horn2013; Graf et al. Reference Graf, Ludin, Arquint, Schmidt, Schaub, Kunz Renggli, Munday, Krezdorn, Baker, Horn, Balmer, Caccone, de Koning and Mäser2016). This highlights an urgent need for new validated drug targets and lead compounds.
Post-translational modifications (PTMs) are covalent and predominantly enzymatic modifications of proteins that occur during or after protein translation. One such PTM is the attachment of lipids (e.g. myristic or palmitic acid) to protein N-termini or side chains. Lipidation is typically catalysed by an acyl transferase that utilizes the coenzyme A (CoA)-activated lipid as a cofactor. In this review we will focus on two protein lipidations: S-acylation, the attachment of a long chain saturated fatty acid (mainly C16:0, palmitate) to a cysteine side chain via a thioester linkage, and N-myristoylation. N-myristoylation is catalysed by an acyl transferase, N-myristoyl transferase (NMT), which attaches myristic acid to the N-terminus of a specific set of protein substrates in lower and higher eukaryotes, thereby forming an amide bond between the 14-carbon saturated fatty acid and an N-terminal glycine (Boutin, Reference Boutin1997). This PTM typically occurs co-translationally (Wilcox et al. Reference Wilcox, Hu and Olson1987) and can be involved in protein–protein interactions, the association of proteins with membranes, and protein stability (Resh, Reference Resh1999, Reference Resh2006; Wright et al. Reference Wright, Heal, Mann and Tate2009). Until recently, relatively little was known about the protein substrates of NMT in protozoan parasites. In P. falciparum, a single NMT isoform was discovered that was shown to myristoylate GAP45 (a protein involved in host cell invasion; Rees-Channer et al. Reference Rees-Channer, Martin, Green, Bowyer, Grainger, Molloy and Holder2006), CDPK1 (life cycle regulation; Möskes et al. Reference Möskes, Burghaus, Wernli, Sauder, Dürrenberger and Kappes2004), ARF1 (trafficking; Leber et al. Reference Leber, Skippen, Fivelman, Bowyer, Cockcroft and Baker2009) and AK2 (energy metabolism; Rahlfs et al. Reference Rahlfs, Koncarevic, Iozef, Mailu, Savvides, Schirmer and Becker2009). In Leishmania, ARL1 (trafficking; Sahin et al. Reference Sahin, Espiau, Tetaud, Cuvillier, Lartigue, Ambit, Robinson and Merlin2008), HASPB (unknown function; Sádlová et al. Reference Sádlová, Price, Smith, Votýpka, Volf and Smith2010) and PPEF (protein phosphatase; Mills et al. Reference Mills, Price, Johner, Emerson and Smith2007) have been reported to be myristoylated, amongst others. Our understanding of the significance of this PTM has been greatly enhanced over the last 5 years by global chemical proteomic strategies, as discussed in the next two sections (‘Approaches for Global Profiling of Protein Lipidation’ and ‘Application of Global Lipidation Mapping Tools in Parasites’). NMT had been identified as a likely essential protein and potential drug target as early as 2000 by Holder et al. for P. falciparum (Gunaratne et al. Reference Gunaratne, Sajid, Ling, Tripathi, Pachebat and Holder2000) and 2003 by Price et al. for L. major and T. brucei (Price et al. Reference Price, Menon, Panethymitaki, Goulding, McKean and Smith2003). Since then, a variety of different small molecule inhibitors have been reported, and recent developments will be discussed in the last section (‘Inhibitors of Protein Lipidation in Protozoan Parasite’).
Chemical proteomic approaches have also recently revealed widespread protein S-acylation in protozoan parasites. Palmitoylacyltransferases (PATs) catalyse S-acylation but the consensus sequence for this modification is even more poorly defined than that for N-myristoylation, and the substrate specificities of the multiple PATs in any one organism are not entirely defined. The dynamic, reversible nature of some S-acylation events makes unravelling the state and function of this modification particularly challenging. PATs are integral membrane proteins with a characteristic DHHC motif within a cysteine-rich domain (CRD) important for catalysis. They localize to membranes of different subcellular compartments via targeting motifs that are not yet understood, and that differ between species. There are 12 PATs in P. falciparum, 11 in the rodent model Plasmodium berghei, and 18 in T. gondii, some of which appear to be essential for parasite survival (Frénal et al. Reference Frénal, Tay, Mueller, Bushell, Jia, Graindorge, Billker, Rayner and Soldati-Favre2013). Furthermore, several recent studies indicate that PATs play stage-specific roles in parasite biology (Beck et al. Reference Beck, Fung, Straub, Coppens, Vashisht, Wohlschlegel and Bradley2013; Hopp et al. Reference Hopp, Balaban, Bushell, Billker, Rayner and Sinnis2016; Santos et al. Reference Santos, Duarte, Kehrer, Ramesar, Avramut, Koster, Dessens, Frischknecht, Chevalley-Maurel, Janse, Franke-Fayard and Mair2016; Tay et al. Reference Tay, Jones, Hodson, Theron, Choudhary and Rayner2016). In trypanosomatids, bioinformatics searches for the DHHC-CRD motif have identified 12 predicted PATs in T. brucei (Emmer et al. Reference Emmer, Souther, Toriello, Olson, Epting and Engman2009), 15 in Trypanosoma cruzi, and 20 in L. major (Goldston et al. Reference Goldston, Sharma, Paul and Engman2014). Interestingly, RNAi knockdown of individual PATs does not affect T. brucei parasite growth in culture, and although this does not exclude a role in virulence or infection; this suggests that there is redundancy and cross-over in PAT substrate specificity (Emmer et al. Reference Emmer, Nakayasu, Souther, Choi, Sobreira, Epting, Nesvizhskii, Almeida and Engman2011). The study of PAT function is hindered by the fact that, in contrast to N-myristoylation, there are no specific chemical inhibitors of PATs. The druggability of these enzymes should be a high priority for further study.
APPROACHES FOR GLOBAL PROFILING OF PROTEIN LIPIDATION
A given PTM is typically of low abundance and often very difficult to detect and quantify directly in the context of a whole proteome. Modified proteins are therefore usually enriched before downstream analysis. However, there are no reliable affinity-based methods to globally enrich lipidated proteins, and historically lipidation has been studied on a protein-by-protein basis using metabolic labelling with poorly sensitive radiolabelled lipid analogues in conjunction with immunoprecipitation. Either specific and highly sensitive antibodies are required, making the approach low throughput, or the protein of interest must be overexpressed, raising questions over the validity of the result in native systems. Computational approaches to predict lipidation of proteins also exist (Maurer-Stroh et al. Reference Maurer-Stroh, Eisenhaber and Eisenhaber2002; Bologna et al. Reference Bologna, Yvon, Duvaud and Veuthey2004; Ren et al. Reference Ren, Wen, Gao, Jin, Xue and Yao2008). However, the sequence motifs are not clearly defined and bioinformatics tools rely on learning sets derived from species such as yeast that may not be transferable to protozoan parasites.
Here we discuss two modern techniques that have been effectively applied in protozoan parasites to globally enrich and identify lipid-modified proteins. The first exploits the chemistry of the PTM linkage, and the second uses tagged lipid analogues that are metabolically incorporated into proteins in the cell. Both approaches have benefited hugely from parallel advances in quantitative proteomics methods and the increasing sensitivity of mass spectrometry (MS) instruments.
Acyl biotin exchange (ABE) chemistry is a well-established technique for detecting S-acylation of proteins (Roth et al. Reference Roth, Wan, Bailey, Sun, Kuchar, Green, Phinney, Yates and Davis2006). ABE is a biotin-switch method that exploits our ability to selectively capture thiols and cleave thioesters to install a biotin affinity tag onto proteins at the site of S-acylation. A protein lysate is treated with a thiol-reactive reagent such as N-ethyl maleimide (NEM) to block free thiols. Subsequently, thioesters (including S-acyl chains) are selectively cleaved with hydroxylamine (HA; Fig. 1). Treatment with the disulphide-forming reagent HPDP-biotin labels the liberated thiols with biotin. A control portion of the lysate is not treated with HA. The two samples are then incubated with an affinity resin (e.g. avidin agarose) and enriched proteins subjected to proteolytic digest. The corresponding peptide fragments are identified by MS-based proteomics and the hits of the two samples are compared. The disulphide linkage between the biotin group and the modified peptide can also be cleaved to enable identification of the S-acylation site. Variations on this technique include acyl-RAC (resin-assisted capture) (Forrester et al. Reference Forrester, Hess, Thompson, Hultman, Moseley, Stamler and Casey2011), where newly exposed thiols derived from thioesters are directly captured on a resin – combining the labelling and enrichment steps – and the recently reported acyl-polyethylene glycol (PEG) exchange (Percher et al. Reference Percher, Ramakrishnan, Thinon, Yuan, Yount and Hang2016), which installs a PEG tag in place of biotin; the mass shifts from the PEG group are readily detectable through gel electrophoresis and Western blot, and can be used to determine levels of S-acylation.
ABE and related approaches are powerful methods for profiling S-acylation. However, ABE provides no information on the nature of the PTM that was incorporated at the thioester site (such as the acyl chain length), is limited to thioester-linked fatty acylations, and cannot distinguish acylation from any other thioester-linked modifications. Incomplete blocking of thiols of abundant proteins can also cause problems of background noise. Metabolic tagging with click chemistry (MTCC) is a complementary approach that is more generally applicable to a variety of lipid modifications (Tate et al. Reference Tate, Kalesh, Lanyon-Hogg, Storck and Thinon2015).
The principle behind MTCC is to use the endogenous machinery of the cell to install a latent chemical tag via the PTM: tagged analogues of the PTM of interest are fed to live cells and incorporated into modified proteins (Fig. 2). The tag must be very small in order to be tolerated by the enzymes that catalyse modification, biorthogonal such that it reacts minimally with the cellular environment, yet reactive enough to act as a chemical handle for downstream capture (with fluorophores or affinity handles such as biotin) and analysis of tagged proteins. The ‘capture’ chemistry most widely used is the copper-catalysed ligation of a terminal alkyne with an azide (a click reaction, also referred to as CuAAC). Although both azido- and alkynyl-fatty acids have been used in metabolic tagging approaches, alkyne-modified lipids are often preferred – mainly due to the empirical observation that this orientation (alkyne on lipid, azide on capture reagent) leads to lower background labelling. MTCC has been applied to detect N-myristoylation and S-palmitoylation of proteins in protozoan parasites, using the tools shown in Fig. 2.
A significant advantage of MTCC is that in principle a tool can be designed to address any PTM in an unbiased way. For example, a myristic acid mimic such as YnMyr (1) or AzMyr (3) (Fig. 2B) could be incorporated onto protein N-termini (as for NMT-catalysed N-myristoylation), or onto other sites, such as S-acylation sites. Tagged analogues should therefore enable detection of less common lipid modifications, such as lysine myristoylation. However, identification of the site of modification can be complex because the biotin–lipid–peptide fragment resulting from protein digest is difficult to detect by MS, or remains anchored to the resin if the proteins are digested on-bead. To tackle this problem, several groups have developed cleavable biotin-azide reagents that allow selective release of the lipidated peptide (e.g. Broncel et al. Reference Broncel, Serwa, Ciepla, Krause, Dallman, Magee and Tate2015). The extent to which lipid analogues are metabolized by the biological system is difficult to assess and remains largely unexplored; this complicates analysis but also provides opportunities to use these tools to map lipid metabolism in diverse systems in the future. As we shall illustrate in the next section, combining ABE and/or MTCC with specific chemical inhibitors or genetic knockdowns of lipid transferase enzymes has proven to be a particularly powerful approach for globally identifying lipidated proteins and assessing the druggability of their cognate transferases.
APPLICATION OF GLOBAL LIPIDATION MAPPING TOOLS IN PARASITES
Over the past 5 years, both ABE and MTCC have been applied in parasites to discover and validate protein lipidation and to probe its inhibition. Prior to the development of these techniques only a handful of proteins had been shown to be lipidated in these organisms, and in many cases only using genetically engineered overexpression systems (reviewed in Tate et al. Reference Tate, Bell, Rackham and Wright2014). Here we discuss recent applications of these techniques in apicomplexan parasites (Plasmodia, Toxoplasma) and in trypanosomatids (Trypanosoma, Leishmania).
Malaria parasites
In 2012, Jones et al. reported the first global study of S-palmitoylation in the asexual stage of the malaria parasite P. falciparum, by applying both MTCC with the well-established palmitate analogue 17-ODYA (4) (Fig. 2) and ABE (Jones et al. Reference Jones, Collins, Goulding, Choudhary and Rayner2012). Quantitative comparison between sample and control in both approaches was carried out using SILAC (stable isotope labelling of amino acids in culture) and the study identified >400 potential palmitoylated proteins. The authors combined ABE with the compound 2-bromopalmitate (2-BP) to analyse the degree to which specific palmitoylations are dynamic. A significant caveat to these results is that 2-BP is not a specific thioesterase inhibitor and has broad non-specific reactivity, including particularly on lipid metabolic pathways (Coleman et al. Reference Coleman, Rao, Fogelsong and Bardes1992; Davda et al. Reference Davda, El Azzouny, Tom, Hernandez, Majmudar, Kennedy and Martin2013; Zheng et al. Reference Zheng, DeRan, Li, Liao, Fukata and Wu2013); the continued use of this molecule despite its potent promiscuity is symptomatic of the lack of well-characterized specific inhibitors for palmitoyl transferases. Despite this issue, the study of Jones et al. was nevertheless a landmark application of ABE and MTCC in a protozoan parasite, and elegantly demonstrates the complementarity of the two techniques (Fig. 3A).
Our laboratory and others have worked extensively on N-myristoylation in malaria and other biological systems, both in terms of inhibitor development (see ‘Inhibitors of Protein Lipidation in Protozoan Parasites’ section) and to globally identify myristoylated proteins. MTCC relies on the cellular machinery to take up fatty acid analogues, convert them into substrates for the acyl transferase (the acyl-CoA thioesters) and incorporate them enzymatically. Early work demonstrated that NMTs will accept azide- and alkyne-tagged myristate mimics in vitro and incorporate them into peptide substrates (Heal et al. Reference Heal, Wickramasinghe, Leatherbarrow and Tate2008). Furthermore, the binding mode of YnMyr-CoA crystallized in the active site of P. vivax NMT (PvNMT) (Fig. 4A) is nearly identical to the conformation adopted by Myr-CoA (Wright et al. Reference Wright, Clough, Rackham, Rangachari, Brannigan, Grainger, Moss, Bottrill, Heal, Broncel, Serwa, Brady, Mann, Leatherbarrow, Tewari, Wilkinson, Holder and Tate2014). MTCC with YnMyr (1) was applied to identify myristoylated proteins in asexual stage P. falciparum schizonts, revealing not only putative N-myristoylated proteins but also proteins known to be modified with a glycosylphosphatidylinositol (GPI) anchor (Fig. 4B). Jones et al. found that 17-ODYA (4) is also incorporated into Plasmodium GPI anchors (Jones et al. Reference Jones, Collins, Goulding, Choudhary and Rayner2012). These results are not surprizing, since Plasmodium GPI anchors are known to incorporate both fatty acids, and illustrate the versatility of the lipid analogues which are readily incorporated by the GPI biosynthetic machinery. N-myristoylation is thought to take place mostly on the N-terminal glycine of substrate proteins. To confirm that YnMyr (1) was attached to these sites, a cleavable azido-biotin reagent (Broncel et al. Reference Broncel, Serwa, Ciepla, Krause, Dallman, Magee and Tate2015) was used: after pull-down of tagged and biotin-labelled proteins, tryptic digest released both the unmodified and modified peptides (Fig. 2A) (Wright et al. Reference Wright, Clough, Rackham, Rangachari, Brannigan, Grainger, Moss, Bottrill, Heal, Broncel, Serwa, Brady, Mann, Leatherbarrow, Tewari, Wilkinson, Holder and Tate2014). Indeed, around 30 modified protein N-termini were detected in this case, providing conclusive evidence for the site-specific attachment of YnMyr (1) to these proteins.
Identifying the mode of action of drugs and small molecules of interest in a live cell context is very challenging, particularly in protozoan parasites. We next exploited our robust and rapid MTCC approach to assess whether NMT inhibitors were acting on-target in the parasite, using both previously reported T. brucei inhibitors (Fig. 10A, compounds 19 and 20) (Frearson et al. Reference Frearson, Brand, McElroy, Cleghorn, Smid, Stojanovski, Price, Guther, Torrie, Robinson, Hallyburton, Mpamhanga, Brannigan, Wilkinson, Hodgkinson, Hui, Qiu, Raimi, van Aalten, Brenk, Gilbert, Read, Fairlamb, Ferguson, Smith and Wyatt2010; Brand et al. Reference Brand, Cleghorn, McElroy, Robinson, Smith, Hallyburton, Harrison, Norcross, Spinks, Bayliss, Norval, Stojanovski, Torrie, Frearson, Brenk, Fairlamb, Ferguson, Read, Wyatt and Gilbert2012) and a novel chemically distinct series developed in-house (Fig. 9A, compounds 15 and analogues) (Rackham et al. Reference Rackham, Brannigan, Rangachari, Meister, Wilkinson, Holder, Leatherbarrow and Tate2014). All five compounds specifically inhibited incorporation of YnMyr (1) into N-myristoylated proteins, and furthermore the in-cell dose–responses calculated from levels of YnMyr (1) incorporation correlated well with EC50 for parasite growth inhibition (Fig. 4C). These experiments therefore demonstrated direct engagement of compounds with NMT in cells and linked parasite death to loss of substrate protein myristoylation. With validated tools in-hand, the phenotype of NMT inhibition in the malaria parasite could be characterized (Wright et al. Reference Wright, Clough, Rackham, Rangachari, Brannigan, Grainger, Moss, Bottrill, Heal, Broncel, Serwa, Brady, Mann, Leatherbarrow, Tewari, Wilkinson, Holder and Tate2014).
YnMyr (1) has proven a versatile tool in Plasmodium species, and this analogue has also been applied in the mouse malaria parasite P. berghei to detect myristoylation of specific proteins involved in sexual development: two inner-membrane complex proteins (Poulin et al. Reference Poulin, Patzewitz, Brady, Silvie, Wright, Ferguson, Wall, Whipple, Guttery, Tate, Wickstead, Holder and Tewari2013), and two protein phosphatases (Guttery et al. Reference Guttery, Poulin, Ramaprasad, Wall, Ferguson, Brady, Patzewitz, Whipple, Straschil, Wright, Mohamed, Radhakrishnan, Arold, Tate, Holder, Wickstead, Pain and Tewari2014).
Toxoplasma gondii
Palmitoylation has been implicated in the intracellular life cycle of the related apicomplexan parasite T. gondii. For example, a palmitoyl protein thioesterase was identified as a target for a small molecule enhancer of host cell invasion, suggesting that dynamic protein S-acylation may play an important regulatory role in this process (Child et al. Reference Child, Hall, Beck, Ofori, Albrow, Garland, Bowyer, Bradley, Powers, Boothroyd, Weerapana and Bogyo2013). Child et al. also used 17-ODYA (4) (Fig. 2B) to verify S-palmitoylation of specific proteins associated with the enhanced invasive phenotype. Building on this work, Foe et al. carried out a global analysis of 17-ODYA (4) tagged proteins in T. gondii extracellular invasive stages (Foe et al. Reference Foe, Child, Majmudar, Krishnamurthy, van der Linden, Ward, Martin and Bogyo2015). Recognizing that the lipid probe may be incorporated into multiple sites in addition to S-palmitoylated cysteines (such as GPI-anchored proteins), the authors also compared samples treated with or without HA. This treatment should selectively cleave thioesters, releasing only S-acylation sites. Quantitative label-free proteomics was used to generate hits within these two experimental set-ups. Comparing the results revealed a final list of ~280 high-confidence S-palmitoylated proteins in T. gondii. Follow-up analysis of one newly identified S-palmitoylated protein, AMA1, which is known to be associated with invasion, showed that one cysteine in particular is likely S-acylated. S-acylation did not appear to be related to AMA1 localization, but a subtle effect on the rate of secretion of microneme proteins was observed. Interestingly, the S-palmitoylated proteome of T. gondii showed limited overlap with palmitoylated orthologues in P. falciparum, perhaps reflecting differences in the life stages analysed, or indicating that most palmitoylation is organism specific (Foe et al. Reference Foe, Child, Majmudar, Krishnamurthy, van der Linden, Ward, Martin and Bogyo2015).
The complementary approach of ABE was also recently applied to T. gondii. Semi-quantitative comparisons of samples treated with and without HA resulted in >400 protein hits (Caballero et al. Reference Caballero, Alonso, Deng, Attias, de Souza and Corvi2016). Around half of these were also identified by Foe et al. using their complementary approach (Fig. 3B). Although only around 50 of the proteins identified by Caballero et al. were found in two biological replicates, suggesting that there is quite some variability in the ABE technique, again around half matched to high-confidence hits from Foe et al. (Supplementary Table S1). Together, these two studies have provided a wealth of data on potential palmitoylated proteins in T. gondii.
Comparisons across the Apicomplexa
Foe et al. compared their T. gondii palmitome dataset (17-ODYA (4)) with the P. falciparum datasets (17-ODYA (4) plus ABE) of Jones et al. by identifying orthologues in these two related species. They noted poor overlap. We added the data of Caballero et al. to this analysis, comparing the aggregate of all three studies: of ~320 putative palmitoylated proteins identified in P. falciparum that have T. gondii orthologues, 141 now have some evidence for S-acylation from either ABE or 17-ODYA (4) analyses in T. gondii (Fig. 3C). As has been noted by the studies described above, it is clear that MTCC and ABE identify not only overlapping but also distinct subsets of the palmitome (and therefore also distinct sets of false positives), and there is value to using both methodologies in combination. In addition, by comparing the P. falciparum data from our myristoylation study with the palmitoylation analyses, we identify 10 likely dually acylated proteins in the parasite (Fig. 3D). The Apicomplexa comparisons are given in Supplementary Table S1, although it should be noted that exact numbers in these comparisons are dependent on how the analysis is carried out: orthologues frequently do not map one-to-one and MS data often cannot distinguish between closely related proteins, which is a widely studied problem in protein inference (Li and Radivojac, Reference Li and Radivojac2012).
Trypanosoma brucei
MTCC was first applied in the sleeping sickness parasite T. brucei to validate the N-myristoylation of a particular protein of interest, ARL6 (Price et al. Reference Price, Hodgkinson, Wright, Tate, Smith, Carrington, Stark and Smith2012), which had a putative role in intracellular protein trafficking. Following metabolic tagging with YnMyr (1) (Fig. 2B), ARL6 was immunoprecipitated from lysate with a specific antibody. Subsequent labelling with a fluorescent azide reagent allowed detection of the modified ARL6 in-gel. Expanding this approach, a global profile of N-myristoylated proteins was performed comparing both bloodstream and insect stages of T. brucei (Fig. 5A) (Wright et al. Reference Wright, Paape, Price, Smith and Tate2016). Out of ~100 robustly enriched proteins in each life stage, roughly half possessed the canonical N-terminal glycine myristoylation motif. Others are known to be GPI anchored or S-acylated, consistent with the frequent observation that S-acylation is more permissive of fatty acid chain length in many eukaryotic systems – i.e. both myristate and palmitate (and their corresponding alkynyl analogues) can be incorporated onto cysteine side chains (Fig. 5B). Indeed, longer chain palmitate analogue YnPal (2) tagged a distinct but overlapping set of proteins in T. brucei (Wright et al. Reference Wright, Paape, Price, Smith and Tate2016). Comparison of the MTCC-derived dataset with the results of an earlier ABE experiment conducted in T. brucei by Emmer et al. (Reference Emmer, Nakayasu, Souther, Choi, Sobreira, Epting, Nesvizhskii, Almeida and Engman2011) revealed some overlap but also differences (Fig. 6A); these are likely the result of both biological (host vs insect form parasites) and technical (MTCC vs ABE) differences in the two studies. Interestingly, YnMyr (1) turned out to be toxic at extended incubation times to bloodstream but not insect forms of T. brucei (Wright et al. Reference Wright, Paape, Price, Smith and Tate2016). This observation is not without precedent (Doering et al. Reference Doering, Lu, Werbovetz, Gokel, Hart, Gordon and Englund1994) and is likely related to effects on the GPI anchor pathway, which is highly dependent on myristate incorporation and crucial for T. brucei host stages (Ferguson et al. Reference Ferguson, Low and Cross1985).
Unlike for S-acylation where multiple, possibly redundant, PAT enzymes exist, NMT appears to be the sole enzyme responsible for protein N-myristoylation. The enzyme has quite narrow substrate specificity for myristoyl-CoA and closely related analogues (Wright et al. Reference Wright, Heal, Mann and Tate2009). Furthermore, whilst there are no specific chemical tools for the inhibition of PATs, for NMT there are several well-characterized molecules available (see ‘Inhibitors of Protein Lipidation in Protozoan Parasite’ section). The T. brucei NMT (TbNMT) inhibitors reported by Frearson et al. (Frearson et al. Reference Frearson, Brand, McElroy, Cleghorn, Smid, Stojanovski, Price, Guther, Torrie, Robinson, Hallyburton, Mpamhanga, Brannigan, Wilkinson, Hodgkinson, Hui, Qiu, Raimi, van Aalten, Brenk, Gilbert, Read, Fairlamb, Ferguson, Smith and Wyatt2010; Brand et al. Reference Brand, Cleghorn, McElroy, Robinson, Smith, Hallyburton, Harrison, Norcross, Spinks, Bayliss, Norval, Stojanovski, Torrie, Frearson, Brenk, Fairlamb, Ferguson, Read, Wyatt and Gilbert2012) specifically reduced YnMyr incorporation into N-myristoylated, but not GPI-anchored proteins in bloodstream form (BSF) parasites (Fig. 5B), demonstrating target engagement in cells. We therefore applied these compounds to simplify the interpretation of the complex YnMyr (1) tagged proteome data, and determine which proteins were true NMT substrates. Parasites were treated with different concentrations of two inhibitors with very different potency, and then proteins tagged with YnMyr (1). After enrichment, quantitative label-free proteomics was used to assess which proteins had reduced YnMyr (1) incorporation in response to inhibition. This analysis revealed ~50 high-confidence NMT substrates; for many of these the YnMyr (1)-modified N-terminal glycine-containing peptide was also identified via cleavable reagents.
Trypanosoma cruzi
NMT is also under investigation as a potential drug target in T. cruzi. Although NMT inhibitors developed against T. brucei were significantly less efficacious in this organism, Roberts et al. showed that the compounds inhibited parasite growth and reduced incorporation of azido-myristate mimic AzMyr (3) (Fig. 2B) in a dose-dependent manner (as read-out by in-gel fluorescence), suggesting that NMT is druggable in this system (Roberts et al. Reference Roberts, Torrie, Wyllie and Fairlamb2014). The authors recently followed this with a study applying AzMyr (3) to identify N-myristoylated proteins in T. cruzi (Roberts and Fairlamb, Reference Roberts and Fairlamb2016). They used both label-free and SILAC quantification and focused on N-myristoylation by treating samples with HA to cleave S-acylation sites. Additionally, they applied two concentrations of their well-characterized NMT inhibitor. This analysis identified ~50 high-confidence N-myristoylated proteins in the parasite. More than half of these had homologues identified as NMT substrates in the T. brucei YnMyr study (Wright et al. Reference Wright, Paape, Price, Smith and Tate2016) (Fig. 6B). Related compounds were also recently shown to act on-target in T. cruzi using a gel-based fluorescent read-out after AzMyr (3) tagging (Herrera et al. Reference Herrera, Brand, Santos, Nohara, Harrison, Norcross, Thompson, Smith, Lema, Varela-Ramirez, Gilbert, Almeida and Maldonado2016).
Leishmania donovani
The extent of protein lipidation in Leishmania species was similarly poorly characterized until recently. We applied YnMyr (1)-based MTCC with label-free quantitative proteomics to assess the potential of NMT as a drug target in L. donovani (Wright et al. Reference Wright, Paape, Storck, Serwa, Smith and Tate2015). As in T. brucei, YnMyr (1) was incorporated into likely N-myristoylated, S-acylated and GPI-anchored proteins, as well as into surface glycolipids that are prevalent in trypanosomatids. A quantitative chemical proteomics-based comparison of YnMyr (1) tagged proteins revealed an overlap of 67% between insect (promastigote) and mammalian host (amastigote) stages of Leishmania parasites, a reflection of their distinct metabolism, and proteome profiles. In addition to enabling study of the different life stages ex vivo, the high sensitivity of MTCC even allowed detection of YnMyr (1) incorporation into native levels of the well-studied N-myristoylated protein HASPB in macrophages infected with amastigotes.
Taking a similar approach to T. brucei, the effects of NMT inhibition on YnMyr (1) modification of each protein were assessed using inhibitor 19 and its N-methylated analogue (19a). These two compounds have nm potency against L. donovani NMT (LdNMT) but dramatically different potencies in cells, with the latter (19a) nearly 50-fold more active against amastigotes than the former (19). Whilst this discrepancy could be due to compound uptake, metabolism or efflux, it also raised the question of whether both compounds were truly acting on-target. YnMyr (1) tagging was performed in the presence of the two compounds at approximately their EC50 values, and revealed the same loss of labelling of specific bands (Fig. 7A). Further quantitative chemical proteomics confirmed this result: YnMyr (1) tagging of the same group of proteins was sensitive to NMT inhibition, demonstrating that both inhibitors engage NMT in live cells. As in other systems, combining chemical inhibition and MTCC proved a powerful approach for dissecting the complex lipidation patterns and ~30 proteins were identified as high-confidence NMT substrates (Fig. 7B). In addition to proteins involved in trafficking, protein phosphorylation, Golgi function and proteasomal degradation, just over half of the hits are completely uncharacterized. Again, there was good overlap with high-confidence T. brucei myristoylated orthologues (Fig. 6C).
Based on this study, it is clear that on-target NMT inhibitors selectively reduce YnMyr (1) incorporation into specific proteins, but do not affect tagging of others (GPI anchored, S-acylated). Indeed, we observed this across Plasmodia, Trypanosoma and Leishmania parasites, as described above. Since YnMyr (1) incorporation can be assessed on-gel, MTCC provides a rapid method to screen for on-target activity of promising NMT inhibitors in these organisms.
Comparisons across the trypanosomatids
The three studies analysing N-myristoylation using MTCC in T. brucei, T. cruzi, and L. donovani, all applied well-characterized inhibitors from the series reported by Frearson et al. in quantitative proteomics experiments to define NMT substrates. This is important because it enables one to distinguish between proteins that incorporate Yn/AzMyr (1/3) at S-palmitoyl or other sites, from those truly N-terminally myristoylated by NMT. With this piece of information and the T. brucei S-palmitoyl proteomics studies of Wright et al. (YnPal MTCC) and Emmer et al. (ABE), candidate proteins for dual acylation in this organism can be identified (Fig. 6D and Supplementary Table S2). This analysis confirms well-validated examples, such as dual acylation of metacaspase 4 and the family of flagellar calcium-binding proteins (Godsel, Reference Godsel1999; Proto et al. Reference Proto, Castanys-Munoz, Black, Tetley, Moss, Juliano, Coombs and Mottram2011), and reveals further avenues of study. For example, the data suggest dual acylation of an ADP-ribosylation factor, two protein phosphatases and numerous proteins involved in fatty acid metabolism, although whether the latter are acylated or bind lipids as part of their catalytic activity remains to be fully explored.
To identify conserved N-myristoylated proteins, the set of high-confidence TbNMT hits were analysed for orthologues in T. cruzi and L. donovani and cross-compared with N-myristoyl datasets from those organisms. There was indeed good overlap between datasets (Fig. 6B and C, Supplementary Table S2), perhaps due to similarity in experimental protocols, as well as the close relatedness of the organisms. The 10 proteins identified across all three organisms (Fig. 6E) are likely only a snapshot of the conserved N-myristoylated proteome, but proteins with functions as diverse as protein degradation (the proteasome subunit), phosphorylation (two protein phosphatases), trafficking (two ARFs), and metabolic regulation (5′AMP-activated protein kinase subunit) can be identified.
INHIBITORS OF PROTEIN LIPIDATION IN PROTOZOAN PARASITES
Considering that acyl transferases catalyse the attachment of modifications that are essential for several vital biological processes in protozoan parasites, the development of corresponding selective and potent small molecule inhibitors could significantly contribute to the limited arsenal of therapies currently available. During the last decade, a variety of different small molecule inhibitors has been identified. The last few years have seen further development of the corresponding scaffolds, as summarized below.
Plasmodium and Leishmania
In order to identify P. falciparum NMT (PfNMT) and PvNMT inhibitor scaffolds, two high-throughput screening (HTS) approaches were performed. For the first HTS a 150 000 compound library was screened with our collaborators at Pfizer in a radioactive scintillation proximity assay (SPA) against PfNMT and LdNMT (Bell et al. Reference Bell, Mills, Williams, Brannigan, Wilkinson, Parkinson, Leatherbarrow, Tate, Holder and Smith2012), while a second HTS was performed in collaboration with MRC Technology (MRCT) and used a 60 000 compound library in a fluorescence-based assay against PvNMT (Goncalves et al. Reference Goncalves, Brannigan, Whalley, Ansell, Saxty, Holder, Wilkinson, Tate and Leatherbarrow2012b ). The fluorescence-based assay format exploits the reaction between the thiol reactive 7-diethylamino-3-(4-maleimido-phenyl)-4-methylcoumarin (CPM) and the free CoA liberated during the enzymatic acyl-transfer (Goncalves et al. Reference Goncalves, Brannigan, Thinon, Olaleye, Serwa, Lanzarone, Wilkinson, Tate and Leatherbarrow2012a ). Both high-throughput screens identified hits in a variety of structural series. SPA-based HTS resulted in the identification of excellent PfNMT inhibitor scaffolds that have been further progressed over the last 4 years. The most promising hit compound of the MRCT HTS was further progressed in a compound series based on a quinoline scaffold (Fig. 8A, compound 5) characterized by micromolar IC50 against PvNMT and moderate selectivity over human NMT (HsNMT) (Goncalves et al. Reference Goncalves, Brannigan, Whalley, Ansell, Saxty, Holder, Wilkinson, Tate and Leatherbarrow2012b ). The HTS of the Pfizer library against LdNMT successfully identified four series (Fig. 8B aminoacylpyrrolidines – compound 7, piperidinylindoles – compound 8, thienopyrimidines and biphenyl derivatives) with good-to-excellent selectivity over all other NMTs tested (Bell et al. Reference Bell, Mills, Williams, Brannigan, Wilkinson, Parkinson, Leatherbarrow, Tate, Holder and Smith2012). The binding mode of all four inhibitor classes was subsequently determined by co-crystallization with LdNMT and/or L. major NMT (LmNMT) (Fig. 8B; Brannigan et al. Reference Brannigan, Roberts, Bell, Hutton, Hodgkinson, Tate, Leatherbarrow, Smith and Wilkinson2014). All inhibitors, apart from the aminoacylpyrrolidines interact via a basic centre with the C-terminal carboxylate of the enzyme. In the case of 7 and 9, the corresponding interaction is mediated by the hydroxyl substituent. Moreover, all compounds show significant interactions with aromatic side chains of Phe90, Tyr217 and Tyr345, and exhibit an additional set of individual interactions. These structural insights were used in a subsequent structure-guided fusion of scaffolds 7 and 8 (Hutton et al. Reference Hutton, Goncalves, Brannigan, Paape, Wright, Waugh, Roberts, Bell, Wilkinson, Smith, Leatherbarrow and Tate2014). The product (Fig. 8B, compound 9) of the inhibitor hybridization exhibits a 40-fold increased potency with good selectivity over HsNMT. However, one major issue with all three inhibitors (7, 8, and 9) is the lack of cell-based activity, which MTCC analysis suggests is due to poor cellular uptake limiting access to NMT in cells (Hutton et al. Reference Hutton, Goncalves, Brannigan, Paape, Wright, Waugh, Roberts, Bell, Wilkinson, Smith, Leatherbarrow and Tate2014; Paape et al. Reference Paape, Bell, Heal, Hutton, Leatherbarrow, Tate and Smith2014).
In parallel an alternative strategy was exploited to identify new PfNMT inhibitors by testing the antimalarial potency of drug molecules that have already been evaluated by pharma companies as lead compounds for the treatment of other diseases. This so called ‘piggyback’ approach was based on a library of 43 inhibitors against NMT from Candida albicans (CaNMT). Although four hits successfully reduced parasitaemia in vitro, the compounds exhibit low ligand efficiency (LE) due to their high molecular weight and high lipophilicity relative to their low enzyme affinity (Bowyer et al. Reference Bowyer, Gunaratne, Grainger, Withers-Martinez, Wickramsinghe, Tate, Leatherbarrow, Brown, Holder and Smith2007, Reference Bowyer, Tate, Leatherbarrow, Holder, Smith and Brown2008). Screening a second small library of 25 inhibitors of CaNMT and TbNMT finally revealed ‘RO-09-4609’ (10) as a moderately selective hit compound. Further optimization resulted in the development of an inhibitor series with a benzo[b]furan scaffold (Fig. 9A, compound 11 + 12) that exhibits a 100-fold affinity improvement over the initial compound. Co-crystallization of inhibitor 12 with PvNMT revealed a competitive binding mode of the benzo[b]furan inhibitors with the peptide substrate (Yu et al. Reference Yu, Brannigan, Moss, Brzozowski, Wilkinson, Holder, Tate and Leatherbarrow2012). The corresponding inhibitors are characterized by moderate enzyme affinity, and the LE was still too low to consider the series to be favourable for further optimization. To overcome this issue, an inhibitor series based on a benzo[b]thiophene scaffold (Fig. 9A, compounds 13–15) was developed to mediate improved π-interactions with two tyrosine residues due to the increased aromatic character of the thiophene moiety. Crystallography of these novel inhibitors with PvNMT revealed an overlapping but distinct binding mode to the benzo[b]furans, with the benzo[b]thiophene moiety being buried deeper within a hydrophobic pocket (Rackham et al. Reference Rackham, Brannigan, Moss, Yu, Wilkinson, Holder, Tate and Leatherbarrow2013). The structure additionally showed that an appropriately positioned methoxyphenyl substituent should be able to interact with Phe105 and Ser319 of PvNMT, thereby increasing the affinity due to further π–π interactions. This hypothesis was validated by increasing the linker length between the benzo[b]thiophene core and the methoxyphenyl substituent. The resulting inhibitor 14 exhibits a 6-fold increased affinity against PfNMT (Fig. 9A; Rackham et al. Reference Rackham, Brannigan, Rangachari, Meister, Wilkinson, Holder, Leatherbarrow and Tate2014). However, one issue with the ester containing benzo[b]thiophenes is the high lipophilic LE (LLE) value of e.g. 13·2 for 13. Highly lipophilic compounds are more likely to partition from plasma to membranes and proteins and thereby exhibit increased promiscuity and toxicity. The LLE value considers lipophilicity, affinity and molecular size. Desirable leads exhibit an LLE of <10 (corresponding to LE > 0·3 and cLogP < 3) (Keserü and Makara, Reference Keserü and Makara2009). To significantly decrease the LLE and to further increase enzyme affinity, 1,3,4-oxadiazole was implemented as a bioisosteric replacement of the ester linker moiety (Rackham et al. Reference Rackham, Brannigan, Rangachari, Meister, Wilkinson, Holder, Leatherbarrow and Tate2014). The corresponding derivatives (Fig. 9A, compound 15) showed a 100-fold improved enzyme affinity and a 100-fold decreased lipophilicity while retaining the selectivity over HsNMT with respect to the first benzo[b]thiophene lead compound 13. In addition to its antiparasitic in vitro activity, compound 15 is potent against four parasite strains, including two drug-resistant ones, and shows promising activity against liver stage (EC50 = 372 nm) parasites.
Scaffold simplification by substituting the bicyclic core with pyridyl (Fig. 5A, compound 16) resulted in the most recently reported oxadiazole-containing inhibitor series (Yu et al. Reference Yu, Brannigan, Rangachari, Heal, Wilkinson, Holder, Leatherbarrow and Tate2015). Remarkably, the scaffold-simplified inhibitors exhibit a similar binding mode in the PvNMT crystal structure as the benzo[b]furan derivatives (Fig. 9A). The 1,2,4-oxadiazole is sandwiched between Y334 and Y211, while the pyridyl nitrogen of 16 additionally stabilizes the enzyme-inhibitor complex via water-mediated hydrogen bonds with Y315. Strikingly, the 3-OMe phenyl moiety of compound 16 also overlays well with the quinoline scaffold of compound 5 (Fig. 8A). Exchanging the trimethylpyrazole with a quinolone moiety finally provided compound 17 (Fig. 9A) with an IC50 of 1·7 nm against PfNMT and good cellular efficacy. The benzo[b]thiophene series was also routinely tested against LdNMT. Remarkably, the affinity spectrum changes significantly if the bicyclic system is truncated to a monocyclic thiophene scaffold (Rackham et al. Reference Rackham, Yu, Brannigan, Heal, Paape, Barker, Wilkinson, Smith, Leatherbarrow and Tate2015). Activity against human and Plasmodium NMTs decreases by almost two orders of magnitude while affinity against LdNMT increases 8-fold. However, since thiophene moieties have been associated with P450 inhibition, a 1,3,4-oxadiazole-containing 5-chlorophenyl derivative was obtained as an optimum scaffold that shows no macrophage toxicity. However, the compound failed to inhibit growth of axenic L. donovani amastigotes (leishmanial stage), likely due to difficulty accessing the target in this parasite. Therefore, further investigation of the physicochemical properties of this series is essential.
Finally, we also reported development of a PvNMT and LmNMT peptidomimetic inhibitor based on an fungal NMT inhibitor (Olaleye et al. Reference Olaleye, Brannigan, Roberts, Leatherbarrow, Wilkinson and Tate2014). The structure of the peptide (Fig. 9B, compound 18) comprizes a Ser-Lys dipeptide, a C-terminal cyclohexyl moiety, and an aliphatic chain at the N-terminus. The resulting peptidomimetic is characterized by sub-micromolar potency against both enzymes and marginal selectivity over HsNMT. Interestingly, 20% of the electron density of the inhibitor-NMT complex structure corresponds to an N-myristoylated inhibitor product and the CoA by-product, providing the first direct structural evidence for a product complex in NMT (Fig. 9B). This complex is presumably formed in situ in the crystal, favoured by high inhibitor occupancy in the solid state.
Toxoplasma
S-palmitoylation is a dynamic PTM that requires a corresponding acyl-protein thioesterase (APT) for the cleavage of the lipid thioesters. In T. gondii, TgPPT1/TgASH1 was recently identified as an orthologue of human APT1. This serine hydrolase can be inhibited by substituted chloroisocoumarin-,β-lactone-, and triazole urea-based inhibitors (Child et al. Reference Child, Hall, Beck, Ofori, Albrow, Garland, Bowyer, Bradley, Powers, Boothroyd, Weerapana and Bogyo2013; Kemp et al. Reference Kemp, Rusch, Adibekian, Bullen, Graindorge, Freymond, Rottmann, Braun-Breton, Baumeister, Porfetye, Vetter, Hedberg and Soldati-Favre2013). Interestingly, these inhibitors enhance tachyzoite invasion. Although enhancers of invasion are not obvious therapeutic agents, Child et al. speculated that the increase in number of host cells infected by multiple parasites and the corresponding increase in the competition for resources within the infected cell might be an unconventional point of action for therapeutics, although further studies are required to test this hypothesis (Child et al. Reference Child, Hall, Beck, Ofori, Albrow, Garland, Bowyer, Bradley, Powers, Boothroyd, Weerapana and Bogyo2013).
2-BP is a small molecule compound that is often incorrectly considered a global inhibitor of palmitoylation. Treatment of T. gondii with 2-BP resulted in altered gliding motility patterns of the parasite and a significant reduction of the invasion process (Alonso et al. Reference Alonso, Coceres, De Napoli, Nieto Guil, Angel and Corvi2012), but as mentioned above these findings should be interpreted carefully, and in full appreciation of the very promiscuous activity and high non-specific toxicity of 2-BP (Davda et al. Reference Davda, El Azzouny, Tom, Hernandez, Majmudar, Kennedy and Martin2013).
Trypanosoma
Pyrazole sulphonamides are NMT inhibitors identified in an HTS against TbNMT by Wyatt et al. (Frearson et al. Reference Frearson, Brand, McElroy, Cleghorn, Smid, Stojanovski, Price, Guther, Torrie, Robinson, Hallyburton, Mpamhanga, Brannigan, Wilkinson, Hodgkinson, Hui, Qiu, Raimi, van Aalten, Brenk, Gilbert, Read, Fairlamb, Ferguson, Smith and Wyatt2010). Eight sulphonamide hits of this initial HTS were further investigated by Maldonado et al. using high content imaging and a metabolic labelling approach. The authors proved the on-target activity of three compounds in sub-micromolar concentrations also against T. cruzi NMT with very low cytotoxic side-effects (Herrera et al. Reference Herrera, Brand, Santos, Nohara, Harrison, Norcross, Thompson, Smith, Lema, Varela-Ramirez, Gilbert, Almeida and Maldonado2016). The lead compound of the HTS, DDD85646 (Fig. 10A, compound 19), shows excellent activity in mouse models during the hemolymphatic peripheral infection stage of T. brucei (stage 1) (Brand et al. Reference Brand, Cleghorn, McElroy, Robinson, Smith, Hallyburton, Harrison, Norcross, Spinks, Bayliss, Norval, Stojanovski, Torrie, Frearson, Brenk, Fairlamb, Ferguson, Read, Wyatt and Gilbert2012). However, due to a low blood–brain barrier (BBB) permeability, the inhibitor is not active in the second stage during which the parasite enters the CNS (central nervous system) thereby giving rise to the classic symptoms of sleeping sickness. Therefore, Read et al. prepared modified pyrazole sulphonamides with a reduced polar surface and a capped sulphonamide group, significantly increasing the BBB permeability (Brand et al. Reference Brand, Norcross, Thompson, Harrison, Smith, Robinson, Torrie, McElroy, Hallyburton, Norval, Scullion, Stojanovski, Simeons, van Aalten, Frearson, Brenk, Fairlamb, Ferguson, Wyatt, Gilbert and Read2014). Their new lead compound (Fig. 10A, compound 20) demonstrated partial efficacy in stage 2 mouse models. However, one issue is that the increased lipophilicity results in off-target effects and poor tolerability of the new lead compound.
Apart from the pyrazole sulphonamides, Gilbert et al. have progressed two further hits from the original HTS and developed a thiazolidinone (e.g. 21) and a benzomorpholinone (e.g. 22) series (Fig. 10A; Spinks et al. Reference Spinks, Smith, Thompson, Robinson, Luksch, Smith, Torrie, McElroy, Stojanovski, Norval, Collie, Hallyburton, Rao, Brand, Brenk, Frearson, Read, Wyatt and Gilbert2015). Like Read et al. the authors aimed at the development of BBB permeable NMT inhibitors. Due to a lack of high-resolution structures of TbNMT and the very high-sequence homology of the binding pockets of TbNMT and LmNMT, the authors used LmNMT as a surrogate. Co-crystallography of the two new TbNMT inhibitor classes with LmNMT revealed that they are characterized by a different binding mode than the sulphonamides (Fig. 10B).
The lead compound of the thiazolidine series (Fig. 10A, compound 21) is characterized by good selectivity over HsNMT, micromolar cellular efficacy, and a good LE value that indicates the potential of the series. The benzomorpholinone series (Fig. 10A, compound 22) contains potent antiparasitic compounds with cellular potencies in the nanomolar range that exhibit BBB permeable compounds. However, the selectivity of the series has to be further improved to enable higher dose levels, and thereby maximizing the chances of curing stage 2 infections.
Concluding remarks
Protein lipidation is an essential PTM for metabolic and cellular processes in protozoan parasites, and its modulation offers interesting opportunities for therapy. Therefore, an extensive investigation of the substrates of protozoan acyl transferases and the corresponding downstream effects of their inhibition is essential. In this context ABE and MTCC are powerful techniques that can be used to profile, image and identify previously unknown lipidated proteins in a data-driven manner and without the need for specific antibodies or protein overexpression. ABE-type approaches can be used on any lysate without the need to optimize incorporation of an analogue and without the risk that the analogue will perturb the system. However, ABE is limited to S-acylation and provides no information on the lipid. MTCC approaches, in contrast, are unbiased and wide in scope. Only proteins dynamically modified during the incubation time with the analogue will be tagged and therefore identified – whilst this can be a potential limitation, more importantly it offers the opportunity for profiling dynamic lipidation through pulse-chase approaches. These technologies have dramatically increased our appreciation of the extent of protein lipidation in parasites and demonstrate that targeting these modifications could have therapeutic value. The identification of small molecule inhibitor scaffolds that inhibit protozoan acyl transferases with high selectivity over the corresponding human enzymes is an important consideration for the development of therapies. In the case of Plasmodium and Leishmania, pyridyl, 1,3,4-oxadiazole containing 5-chlorophenyl, and peptidomimetic-based scaffolds show promising characteristics and good cellular efficacy, including against liver stage malaria parasites. In the case of Trypanosoma, pyrazole sulphonamides, thiazolidinone and benzomorpholinone inhibitors are potent antiparasitic compounds; however, BBB permeability and selectivity needs to be further improved to increase their efficacy against stage 2 of T. brucei infection. Bringing together lipid profiling technologies and medicinal chemistry efforts, MTCC platforms in particular have been successfully used to demonstrate the on-target mode of action of NMT inhibitors in live parasites.
Evidence from analytical tools has accumulated to the point where S-acylation must be considered a major regulatory pathway in all eukaryotes (Resh, Reference Resh2016). The enzymes involved in removal of this modification come from the superfamily of serine hydrolases, and selective small molecule inhibitors are available for some of these enzymes. Their inhibition can lead to interesting and unexpected phenotypes, as mentioned above, and further characterization of their apparently broad substrate scope and complex localization will be important in validating them as potential drug targets. In contrast, the diverse class of protein S-palmitoyl transferases (PATs), including >20 genes in humans, has yet to yield to small molecule inhibitor discovery, and the chemical tools available for PATs are effectively non-existent. Indeed, the continued use of 2-BP due to its commercial availability is to greatly compound the challenges of the field due to the exceptional promiscuity of this molecule, as noted above. Robust and widely applicable CRIPSR-Cas gene-knockout approaches will be an important enabling tool to unpick the roles of PATs in parasites and in the host, but the discovery of cell-active inhibitors selective for the class, or for members of the class, would be transformative for the field, and should be pursued as a high priority.
In contrast, the scope for NMT as a target in eukaryotic pathogens is very clear, and may be very broad, as recently demonstrated for helminths (Galvin et al. Reference Galvin, Li, Villemaine, Poole, Chapman, Pollastri, Wyatt and Carlow2014). The availability of multiple potent inhibitor series and powerful tools to analyse PTMs in living systems greatly enhances the opportunities for drug development against this target. With the exception of T. brucei, which is rapidly killed by NMT inhibition due to its exceptional reliance on myristoylation-dependent trafficking, NMT inhibition has quite an extended mode of action. This is hypothesized to be due to an indirect dependence on protein degradation: myristoylated proteins that were present prior to inhibition will typically need to undergo some degree of degradation in order for inhibition of co-translational myristoylation to impact viability. Careful consideration of compound uptake in cells, distribution/pharmacokinetics and pharmacodynamics (the dynamics of target engagement) will be required to realize the potential of NMT inhibitors as antiparasitic agents, and research towards this objective continues in our laboratories, in collaboration with other research groups.
SUPPLEMENTARY MATERIAL
The supplementary material for this article can be found at https://doi.org/10.1017/S0031182017000282.
ACKNOWLEDGEMENTS
The authors are grateful to the anonymous reviewers of this manuscript for suggesting the detailed and informative analyses presented in Figs 3 and 6 and Supplementary Tables 1 & 2.
FINANCIAL SUPPORT
Financial support by Medicines for Malaria Venture (MMV) is gratefully acknowledged, and from the Engineering and Physical Sciences Research Council (EPSRC, grant EP/F500416/1).