Submit manuscript...
MOJ
eISSN: 2374-6920

Proteomics & Bioinformatics

Correspondence:

Received: January 01, 1970 | Published: ,

Citation: DOI:

Download PDF

Abstrat

Chemical biology approaches combined with proteomics provide a powerful tool to dissect biological mechanisms with high precision, accuracy and temporal resolution. In the recent years, infectious disease biology, in particular, translational research to discover new drug targets for human diseases has benefited immensely by leveraging this approach. Identification of novel drug targets is critical to combat infections, such as human malaria. Malaria parasites tend to acquire drug resistance very rapidly warranting the design of new lines of chemotherapy. In this article, we briefly review recent discoveries in the field of malaria research that utilizes chemical biology and proteomics targeted at identifying viable drug scenarios and potential drug targets.

Keywords: malaria, chemical biology, activity-based probes, proteomics, target identification, drug discovery, plasmodium falciparum

Abbreviations

ABP, activity-based probe; MS, mass spectrometry; P. vivax, plasmodium vivax

Introduction

Malaria is among the deadliest and wide-spread infectious diseases. Although, there has been significant progress in malaria prevention and control through chemotherapeutic approaches and vector control initiatives, half of the world’s population still inhabits in malaria endemic regions.1 The situation is even more alarming due to the emergence of rapid parasitic resistance against commonly used drugs.2–5 Hence it has become all the more important to design new ways of tackling this deadly infection. Among several variants of plasmodial parasites infecting humans, the most serious pathological condition is caused by Plasmodium falciparum infections. Asexual intra-erythrocytic developmental processes, primarily steps associated with hemoglobin degradation, parasite invasion or parasite release are known to be viable checkpoints for potential chemotherapy development. Identification and detailed exploration of such scenarios requires highly robust and easily adaptable methodologies. Functional proteomics approaches using small molecule inhibitors and Activity-Based Probes (ABPs), present an ideal platform to temporally dissect essential parasitic survival mechanisms against which chemotherapy may be developed.

Discussion

Among the many antimalarial drugs currently used, quinolones and artesunate constitute the most widely used and effective.6,7 However, due to the emergence of resistant forms of parasites against these commonly used drugs, identification of novel drug targets is urgently warranted.8,9 During the intra-erythrocytic developmental cycle, plasmodial parasites engage in highly regulated interactions with its host cells to facilitate entry, survival, replication and eventual release. Detailed understanding of these life-stage events using genetic, biochemical or chemical tools will provide new avenues for potential chemotherapy development against malaria. Such processes during the developmental cycle that lasts about 48 hours are unique and are mediated by specific sets of effector proteins that are either expressed or activated only at the required life stages. Most of these proteins are essential for parasitic survival and cannot be deleted genetically limiting the application of genetic manipulation techniques to study them in greater detail.

In contrast, small molecule inhibitors capable of altering a biological pathway without interfering with transcriptional or translational machineries provide an efficient tool to (a) selectively manipulate the biological event, (b) generate chemical phenotypes that can be studied using alternate methods and (c) directly identify/validate the target molecules that orchestrate these processes. To do this, small molecule libraries that are designed to influence the desired life-stage are tested on cultured parasites to evaluate their efficacy and to identify possible phenotypes. Once phenotypic characterization is completed, chemically modified forms of the potent scaffolds are developed as activity-based probes (ABPs). These probes maintain the active functional groups required for its inhibitory potential, however, also carry additional reporter moieties to enable functional read outs of inhibition, linked through appropriate chemical linkers (Figure 1). These reporter moieties aid monitoring of the sub-cellular targets in real time. Although, fluorophores such as BODIPY or TAMRA10–12 are used widely as reporters, biotinylated molecules13 are the most preferred as they allow the affinity purification/identification of the targets using streptavidin-based affinity purification methods. Such approaches have widely been used in the study of malaria biology, in relevance to host invasion, parasitic progression/replication as well as merozoite egress as represented in Figure 1. This review describes the novel drug targets unraveled through such approaches.

Figure 1 Flow chart describing the application of Activity-based Probes to identify novel antimalarial drug targets during parasite invasion, progression or egress from infected cells. ABPs allow selective manipulation of a biochemical cascade and detailed dissection of the impaired life-stage event and to eventually identify viable drug targets.

Drug targets relevant to invasion

Red blood cell invasion by plasmodial parasites is rapid and often completed in less than a minute. The process consists of distinct interaction steps between invading merozoites and their host cell surface in a concerted effort of sugar-binding proteins, adhesins and proteases. Among the several plasmodial surface proteins, MSP-1 is the most abundant protein on the merozoite surface14–16 and is known to be one of the primary mediators of merozoite invasion. It is synthesized as precursor protein, which undergoes proteolytic processing during merozoite egress. During invasion, this complex is further fragmented and only its C-terminal portion remain attached to the parasite surface whereas the remaining portions are shed off. Antibodies targeting various domains of MSP-1 have been shown to effectively block invasion,17 although they lack therapeutic potential due to poor permeability. Hence, inhibitors interfering with MSP-1 processing represent highly promising drug candidates.18,19 A number of sugar derivatives have been shown to bind to MSP-1 and block invasion, highlighting an opportunity to target this protein for antimalarial development. Recently, heparin was shown to inhibit MSP-1 processing by binding to one of its subunit called MSP-133, thereby inhibiting invasion. Using affinity purification approaches, Boyle et al.,20 were able to demonstrate specific binding to heparin of MSP-133, and not MSP-119 to heparin. More recently, combining chemical biology, computational biology and biochemistry, our group described the identification of a small molecule, NIC that was able to bind to the shorter C-terminal fragment of MSP-1 (MSP-119). Small molecules evaluated in this study were initially selected through virtual screening of compounds with potential binding affinity to MSP-119. Subsequently, detailed chemical biology experiments in combination with biochemical as well as proteomics-based validation confirmed specific and direct binding of NIC to plasmodia MSP-119. Our results revealed that binding of NIC to MSP-119 resulted in reduced invasiveness of merozoites derived from P. falciparum and P. vivax.21 These results represent a major advancement in the field of antimalarial development targeted against invasion proteins, since the residues within MSP-119 that binds to NIC are significantly conserved across multiple plasmodia, highlighting the possibility to explore such scaffolds for pan-antimalarial development.

Parasite invasion also involves shedding and presentation of surface proteins such as MSP-1 and AMA-1,22,23 which appears to be driven by serine proteases of the subtilisin family. Among such enzymes, Subtilisin-1 like protease (Sub-1) was shown to be proteolytically active during both invasion and egress. Sub-1 is capable of processing a variety of parasite surface proteins such as MSP1/6/7 and members of a family of papain-like putative proteases called SERA,24 thereby aiding invasion and egress.25,26 Recent research demonstrates the targeted development of novel α-ketoamide based molecules as well as quinolylhydrazones27 as powerful inhibitors of this enzyme. Another much studied sheddase; subtilisin-2 like protease of the malaria parasite appears to process invasion proteins such as AMA-1.28 The important cysteine proteases such as falcipain-2 and falcipain-3 are validated as potential targets upon which innovative classes of new drugs are being designed.29–31

Rupture mediators as drug targets

Application of chemical biology and proteomics has played an important role to discover various parasitic and host proteases involved in host cell rupture thereby releasing plasmodial merozoites. Blocking the activation of such proteases irreversibly locks mature merozoites within the host cells preventing a further cycle of infection. Serine Repeat Antigens (SERAs) play a critical role in egress pathway32 and this in turn is facilitated by upstream active proteases such as PfSUB133 and DPAP3. Inhibitors of DPAP3 and PfSUB1 blocked the processing of SERAs 5 & 6 thereby blocking egress,34 showcasing the role of these proteases in triggering host cell collapse and thus, their potential as drug targets. In addition, studies using inhibitors of papain family cysteine proteases and corresponding ABPs have demonstrated the importance of host calpain activation during latter stages of parasite development35 in Plasmodium. Calpain activation resulted in irreversible cytoskeletal damage in the erythrocyte and thereby aided parasite release.36 Furthermore, this study provided direct evidences depicting both Plasmodium and Toxoplasma confiscates calpain proteases to facilitate parasite egress and that silencing host calpain-1 activity, either biochemically or genetically, resulted in impaired parasite exit. Development of host calpain specific inhibitors will be a new avenue for antimalarial development leveraging on host cell pathways required for parasitic replication.35,37,38

Other emerging targets

Digestive vacuole enzymes have been extensively explored as candidate drug targets against malaria.39 Recent investigations also highlight the possibility to approach a number of other proteins critical for parasite survival and replication as viable targets for novel chemotherapy development. For instance, spiroindolones that targets a P-type cation-transporter ATPase 4 (PfATP4) was shown to be effective against P. falciparum and P. vivax malaria40,41 and compounds of that lineage are currently undergoing clinical validation. N-myristoyltransferase, an enzyme required for the myristoylation of several plasmodial proteins appears to be a promising target.42 Targeting this enzyme using selective inhibitors irreversibly damaged the assembly of inner membrane complex thereby impairing parasite replication both in vitro and in vivo. Another emerging candidate target is plasmodial signal peptide peptidase, a protease component of ER-associated degradation (ERAD)43 pathway, required for the degradation of unstable proteins.44 Although research pertaining to these novel targets is still evolving, these recent trends highlight the importance of chemical biology approaches in novel target hunting.

Conclusion

The development and design of new antimalarial drugs that are able to prevent invasion, rupture and other essential parasitic pathways will provide unprecedented advantage to combat the deadly infection of malaria. To facilitate the design of novel and efficient antimalarial drugs, it is important to understand parasitic genetics and resistance mechanisms in greater depth and resolution. Where employing classical genetic approaches has practical limitations in studying malaria, chemical biology combined to proteomics and biochemistry provide a suitable platform to probe hitherto unexplored molecular mechanisms and protein targets with great precision that can be exploited for future antimalarial design.

Acknowledgements

The authors acknowledge the following grants: SRG LSC 2013 049 (RC) and ZJURP1300102 (RC, AS and CTTC) by Singapore University of Technology. GS acknowledges President’s Graduate Fellowship awarded by the Ministry of Education, Singapore.

Conflict of interest

The author declares no conflict of interest.

References

  1. http://www.who.int/malaria/world_malaria_report_2011/en/
  2. Kaay H. Chemotherapy and drug resistance in malaria – Peters W. Tropical and Geographical Medicine. 1971;23:309–310.
  3. Mita T, Tanabe K, Kita K. Spread and evolution of Plasmodium falciparum drug resistance. Parasitol Int. 2009;58(3):201–209.
  4. O'Brien C, Henrich PP, Passi N, et al. Recent clinical and molecular insights into emerging artemisinin resistance in Plasmodium falciparum. Curr Opin Infect Dis. 2011;24(6):570–577.
  5. Thanh NV, Toan TQ, Cowman AF, et al. Monitoring for Plasmodium falciparum drug resistance to artemisinin and artesunate in Binh Phuoc Province, Vietnam:1998–2009. Malaria J. 2010;9:181.
  6. Thomé R, Lopes SC, Costa FT, et al. Chloroquine: modes of action of an undervalued drug. Immunol Lett. 2013;153(1–2):50–57.
  7. Banek K, Lalani M, Staedke SG, et al. Adherence to artemisinin–based combination therapy for the treatment of malaria: a systematic review of the evidence. Malaria Journal. 2014;13(1):7.
  8. Wongsrichanalai C, Pickard AL, Wernsdorfer WH, et al. Epidemiology of drug–resistant malaria. Lancet Infect Dis. 2002;2(4):209–218.
  9. Guiguemde WA, Shelat AA, Garcia–Bustos JF, et al. Global Phenotypic Screening for Antimalarials. Chem Biol. 2012;19(1):116–129.
  10. Enkvist E, Viht K, Bischoff N, et al. A subnanomolar fluorescent probe for protein kinase CK2 interaction studies. Org Biomol Chem. 2012;10(43):8645–8653.  
  11. Fu Y, Klonis N, Suarna C, et al. A Phosphatidylcholine–BODIPY 581/591 Conjugate Allows Mapping of Oxidative Stress in P. falciparum–Infected Erythrocytes. Cytometry A. 2009;75(5):390–404.
  12. Lin W, Liu J, Jeffries C, et al. Development of BODIPY FL Vindoline as a Novel and High–Affinity Pregnane X Receptor Fluorescent Probe. Bioconjug Chem. 2014;25(9):1664–1677.
  13. Greenbaum D, Baruch A, Hayrapetian L, et al. Chemical approaches for functionally probing the proteome. Mol Cell Proteomics. 2002;1(1):60–68.
  14. Dvorak JA, Miller LH, Whitehouse WC, et al. Invasion of erythrocytes by malaria merozoites. Science. 1975;187(4178):748–750.
  15. Child MA, Epp C, Bujard H, et al. Regulated maturation of malaria merozoite surface protein–1 is essential for parasite growth. Mol Microbiol. 2010;78(1):187–202.
  16. Holder AA, Blackman MJ, Burghaus PA, et al. A malaria merozoite surface protein (MSP1)–structure, processing and function. Mem Inst Oswaldo Cruz. 1992;87 (Suppl 3):37–42.
  17. Moss DK, Remarque EJ, Faber BW, et al. Plasmodium falciparum 19–kilodalton merozoite surface protein 1 (MSP1)–specific antibodies that interfere with parasite growth in vitro can inhibit MSP1 processing, merozoite invasion, and intracellular parasite development. Infect Immun. 2012;80(3):1280–1287.
  18. Carruthers VB, Blackman MJ. A new release on life: emerging concepts in proteolysis and parasite invasion. Molecular Microbiology. 2005;55(6):1617–1630.
  19. Fleck SL, Birdsall B, Babon J, et al. Suramin and suramin analogues inhibit merozoite surface protein–1 secondary processing and erythrocyte invasion by the malaria parasite Plasmodium falciparum. J of Biol Chem. 2003;278(48):47670–47677.
  20. Boyle MJ, Richards JS, Gilson PR, et al. Interactions with heparin–like molecules during erythrocyte invasion by Plasmodium falciparum merozoites. Blood. 2010;115(22):4559–4568.
  21. Chandramohanadas, Basappa, Russell B, et al. Small Molecule Targeting Malaria Merozoite Surface Protein–1 (MSP–1) Prevents Host Invasion of Divergent Plasmodial Species. J Infect Dis. 2014;210(10):1616–1626.
  22. Triglia T, Healer J, Caruana SR, et al. Apical membrane antigen 1 plays a central role in erythrocyte invasion by Plasmodium species. Mol Microbiol. 2000;38(4):706–718.
  23. Woehlbier U, Epp C, Hackett F, et al. Antibodies against multiple merozoite surface antigens of the human malaria parasite Plasmodium falciparum inhibit parasite maturation and red blood cell invasion. Malar J. 2010;9:77.
  24. De Monerri NCS, Flynn HR, Campos MG, et al. Global Identification of Multiple Substrates for Plasmodium falciparum SUB1, an Essential Malarial Processing Protease. Infect Immun. 2011;79(3):1086–1097.
  25. Uzureau P, Barale JC, Janse CJ, et al. Gene targeting demonstrates that the Plasmodium berghei subtilisin PbSUB2 is essential for red cell invasion and reveals spontaneous genetic recombination events. Cell Microbiol. 2004;6(1):65–78.
  26. Withers–Martinez C, Jean L, Blackman MJ. Subtilisin–like proteases of the malaria parasite. Mol Microbiol. 2004;53(1):55–63.
  27. Kher SS, Penzo M, Fulle S, et al. Substrate derived peptidic alpha–ketoamides as inhibitors of the malarial protease PfSUB1. Bioorg Med Chem Lett. 2014;24(18):4486–4489.
  28. Harris PK, Yeoh S, Dluzewski AR, et al. Molecular identification of a malaria merozoite surface sheddase. PLoS Pathog. 2005;1(3):241–251.
  29. Greenbaum DC, Baruch A, Grainger M, et al. A role for the protease falcipain 1 in host cell invasion by the human malaria parasite. Science. 2002;298 (5600):2002–2006.
  30. Marques AF, Esser D, Rosenthal PJ, et al. Falcipain–2 inhibition by suramin and suramin analogues. Bioorg Med Chem. 2013;21(13):3667–3673.
  31. Rosenthal PJ. Falcipains and other cysteine proteases of malaria parasites. Adv Exp Med Biol. 2011;712:30–48.
  32. Ruecker A, Shea M, Hackett F, et al. Proteolytic activation of the essential parasitophorous vacuole cysteine protease SERA6 accompanies malaria parasite egress from its host erythrocyte. J Biol Chem. 2012;287(45):37949–37963.
  33. Yeoh S, O'Donnell RA, Koussis K, et al. Subcellular discharge of a serine protease mediates release of invasive malaria parasites from host erythrocytes. Cell. 2007;131(6):1072–1083.
  34. Arastu–Kapur, Ponder EL, Fonović UP, et al. Identification of proteases that regulate erythrocyte rupture by the malaria parasite Plasmodium falciparum. Nat Chem Biol. 2008;4(3):203–213.
  35. Chandramohanadas R, Davis PH, Beiting DP, et al. Apicomplexan Parasites Co–Opt Host Calpains to Facilitate Their Escape from Infected Cells. Science. 2009;324(5928):794–797.
  36. Millholland MG, Chandramohanadas R, Pizzarro A, et al. The Malaria Parasite Progressively Dismantles the Host Erythrocyte Cytoskeleton for Efficient Egress. Mol Cell Proteomics. 2011:10(12).
  37. Rahul CN, Shiva Krishna K, Pawar AP, et al. Genetic and structural characterization of PvSERA4: potential implication as therapeutic target for Plasmodium vivax malaria. J Biomol Struct Dyn. 2014;32(4):580–590.
  38. Palacpac NMQ, Leung BWY, Arisue N, et al. Plasmodium vivax serine repeat antigen (SERA) multigene family exhibits similar expression patterns in independent infections. Mol Biochem Parasitol. 2006;150(2):353–358.
  39. Teixeira C, Gomes JR, Gomes P. Falcipains, Plasmodium falciparum cysteine proteases as key drug targets against malaria. Curr Med Chem. 2011;18(10):1555–1572.
  40. Spillman NJ, Allen RJ, McNamara CW, et al. Na+ Regulation in the Malaria Parasite Plasmodium falciparum Involves the Cation ATPase PfATP4 and Is a Target of the Spiroindolone Antimalarials. Cell Host Microbe. 2013;13(2):227–237.
  41. Yeung BKS, Zou B, Rottmann M, et al. Spirotetrahydro beta–Carbolines (Spiroindolones): A New Class of Potent and Orally Efficacious Compounds for the Treatment of Malaria. J Med Chem. 2010;53(14):5155–5164.
  42. Wright MH, Clough B, Rackham MD, et al. Validation of N–myristoyltransferase as an antimalarial drug target using an integrated chemical biology approach. Nat Chem. 2014;6(2):112–121.
  43. Harbut MB, Patel BA, Yeung BK, et al. Targeting the ERAD pathway via inhibition of signal peptide peptidase for antiparasiticsd therapeutic design. Proc Natl Acad Sci U S A. 2012;109(52):21486–21491.
  44. Smith MH, Ploegh HL, Weissman JS. Road to Ruin: Targeting Proteins for Degradation in the Endoplasmic Reticulum. Science. 2011;334(6059):1086–1090.
Creative Commons Attribution License

© . This is an open access article distributed under the terms of the, which permits unrestricted use, distribution, and build upon your work non-commercially.