1,798
Views
2
CrossRef citations to date
0
Altmetric
Editorial

How can proteomics overhaul our understanding of Leishmania biology?

&
Pages 789-792 | Received 30 Nov 2020, Accepted 01 Feb 2021, Published online: 10 Feb 2021

1. Introduction

In the post-genomic era, genomic and transcriptomic techniques have significantly advanced our understanding of the fundamental genetic aspects of many human diseases. However, these techniques have a narrow scope in the study of cellular processes of disease mechanisms. For instance, gene- or transcript-based studies are totally blind to the profound implications of post-translational modifications (PTMs) in normal cellular physiology and in pathology. Events such as alternative splicing, somatic recombination, proteolytic cleavage, single amino acid polymorphisms and PTMs could lead to over 100 protein isoforms per gene [Citation1]. More than 95% of currently known drug targets are proteins, of which about 55% are enzymes [Citation2]. The functional expression of proteins in cells and tissues and the catalytic activity of enzymes are modulated by various molecular mechanisms that operate at the protein level – such as protein-protein interactions, co- and post-translational modifications and cellular localization, on which genomic and transcriptomic methods can provide very little to no information. As proteins serve as the work horses of the cell, proteomics, the large-scale study of proteins in an organism, has recently gained significant traction in the biological sciences as a powerful methodology for molecular-level characterization of the processes of life and disease development.

Proteomics has a special place in the study of the protozoan parasites Leishmania spp., the etiological agents of the complex Neglected Tropical Disease (NTD) leishmaniasis. The disease is endemic in over 90 countries in the tropics, sub-tropics, and southern Europe, affecting the lives of over 200 million people with about 1.5 million new cases and an estimated 20,000 to 50,000 deaths annually [Citation3]. At least 20 different species of the genus Leishmania have been identified as causing disease in humans, with distinct species leading to different clinical manifestations ranging from various forms of cutaneous lesions to the deadly visceral form. An ancient lineage in the evolution of eukaryotes unicellular Leishmania spp., unlike higher eukaryotes, lack promoter-mediated gene expression and regulation. Instead, they rely on post-transcriptional mechanisms such as mRNA processing, translation, and degradation for regulating nuclear gene expression [Citation4]. Proteins are at the centre of all these crucial processes and recent developments in proteomic technologies have the potential to transform our understanding of the biology of Leishmania spp. Herein we discuss the unique strengths and scope of some of these proteomic methodologies.

2. The curious case of life cycle differentiation, stress adaptation, and PTMs of Leishmania

During their life cycle Leishmania spp. undergo several morphological transformations, ranging from the multiple promastigote forms in the phlebotomine sandfly host to an amastigote form in the mammalian host. These transformations reflect the adaptation of the parasite to the environmental changes experienced in the vertebrate and invertebrate hosts. Characterizing the underlying biochemical changes in the parasite during this differentiation may provide valuable information for antileishmanial drug development strategies. A handful of proteomic mass spectrometry (MS) studies that compared the proteomes of the different life cycle stages of the Leishmania parasites have provided information on many differentially expressed proteins [Citation5]. However, further studies are required to establish whether the observed differences are the cause or the effect of the life cycle differentiation. Similarly, the extraordinary ability of Leishmania spp. to adapt to environmental stresses also calls for in-depth study. Molecular-level characterization of stress adaptation in Leishmania spp. will shed light into not only the unknown biology of the parasite, but also into the mechanisms of drug resistance. Using a combination of BONCAT (bioorthogonal noncanonical amino acid tagging) metabolic labeling and quantitative proteomic MS, we recently showed that whilst nutrient deprivation causes a repression of global protein synthesis in L. mexicana, a set of starvation-responsive proteins are preferentially synthesized [Citation6]. In principle, the BONCAT-quantitative proteomic MS combination could be employed for accurate measurement of perturbations in the Leishmania proteome induced by a variety of stress conditions. Although highly powerful, the method provides measurement of only the newly synthesized proteome (NSP). Recent developments in proteomic MS technologies, such as the mPDP (multiplexed proteome dynamic profiling) [Citation7] that exploits a combination of dynamic SILAC (stable isotope labeling by amino acids in cell culture) labeling and isobaric mass tagging, have the potential to simultaneously profile the effects of perturbing conditions on both the mature (preexisting) proteome and the NSP.

In addition to changes in protein abundance, stage-specific phosphorylation events have been reported in Leishmania spp. [Citation8], pointing to functional regulatory role for phosphorylation in the organism. The presence of many protein kinases and protein phosphatases in the Leishmania genome hints the potential role of reversible protein phosphorylation on modulating protein activity and cell signaling in the organism. Glycosylation [Citation9,Citation10], methylation [Citation10], and acetylation [Citation10] were also reported in Leishmania spp. Similarly, bioinformatics has revealed that the Leishmania genome harbors several E1, E2, and E3 ligases as well as many deubiquitinating cysteine peptidases (DUBs) [Citation11]. Using activity-based protein profiling and MS, a recent study identified six active DUBs in L. mexicana [Citation12] providing the first protein-level evidence for active DUBs in Leishmania spp. Also, in L. mexicana 28 enzymes of the ubiquitination pathway have recently been characterized, of which many are found to play important roles in life cycle progression or in vivo infection [Citation11]. In the absence of transcriptional regulation in Leishmania spp., it is likely that the different PTMs serve crucial roles in differentiation, stress adaptation, and interaction with the host. At present, however, the study of PTMs is underdeveloped in the Leishmania spp. Large-scale proteomic MS discovery studies are required to identify the complete repertoire of PTMs, establish the cross-talks between the different PTMs and to elucidate their biological functions in the organism.

In addition to profiling PTMs, our ability to accurately determine the localization of proteins within the different specialized cell compartments and structures in the parasite will provide valuable information on the protein function and the elusive biology of Leishmania spp. Toward this, microscopy methods are typically employed. Although highly useful, these are limited by low-throughput and availability of specific fluorescent reporters of individual proteins. Although not yet reported in Leishmania spp, the spatial proteomic MS technology hyperLOPIT (hyperplexed localization of organelle proteins by isotope tagging) [Citation13] holds great potential for high-throughput characterization of steady-state subcellular localization of Leishmania proteins and may provide a comprehensive understanding of the proteomic organization in the parasite as recently reported in the case of Toxoplasma gondii [Citation14].

3. Characterizing Leishmania-host interactions

The development and severity of clinical manifestations of leishmaniasis depend not only on the Leishmania spp. but on the many factors pertaining to the susceptible individual such as malnutrition, comorbidities and the state of the immune system. Leishmania spp. parasites have co-evolved with humans in endemic areas and this positive selection pressure has contributed to pathogen survival by latent infection. The development of post-Kala-azar dermal leishmaniasis (PKDL) in some visceral leishmaniasis (VL) patients after recovery from the VL, and the development of mucosal lesions in some individuals after several years or even decades of developing primary cutaneous lesions, are indicative of the ability of the parasite to persist within the host even after successful treatment of the initial clinical condition. Clearly, despite years of research, many aspects of the Leishmania-host interaction remain poorly understood. In principle, all antileishmanials that work purely by targeting a parasite protein are likely to eventually fail as the extraordinary genetic plasticity of Leishmania spp. will confer fitness gains enabling the parasite to effectively evolve toward drug-resistant phenotypes. Therefore, an alternative strategy of host-directed therapeutic development has been proposed to tackle this issue. However, in the first place this requires better understanding of the Leishmania-host interaction.

Proteomic MS has enabled detection of changes in the host protein expression profiles in macrophage in vitro infection models [Citation15,Citation16], murine and human cutaneous lesions [Citation17,Citation18] and in the serum of infected individuals [Citation19]. Proteomic MS studies have demonstrated that the Leishmania infection causes a species-specific reprogramming of major metabolic pathways in the host cell [Citation16]. Similarly, Leishmania spp. may cause epigenetic reprogramming in the host cell by altering DNA methylation or histone post-translational modifications [Citation20]. However, further studies are required to profile and understand these processes in greater depth. It is anticipated that in the coming years proteomic technologies will shed more light on the molecular interactions and impact of intracellular Leishmania spp. infection on the host cell.

4. Chemical proteomics and thermal proteome profiling

The use of synthetic chemical probes in combination with proteomic MS, known as chemical proteomics, has emerged as a powerful approach for unraveling protein function in various biological systems. Chemical proteomics workflows typically rely on a suitable quantitative proteomic method such as SILAC, iTRAQ (isobaric tags for relative and absolute quantitation) or TMT (tandem mass tag) labeling or LFQ (label-free quantification) for quantitative comparison of the different conditions of the study. Chemical proteomics in Leishmania spp., although in its infancy, has showed glimpses of its unique capabilities. For instance, in a seminal study, Wyllie et al. employed SILAC-based chemical proteomics to identify a set of L. donovani cyclin-dependent kinases (CDKs) as molecular targets of their antileishmanial preclinical candidate compound [Citation21]. Another noteworthy study involves global profiling of substrates of L. donovani N-myristoyltransferase (NMT) using metabolic incorporation with a clickable myristic acid analog and LFQ proteomic MS [Citation22]. With further development in the chemical probing of specific Leishmania proteins, the field of chemical proteomics could significantly contribute to increasing our understanding of the parasite’s biology.

Whilst chemical proteomics requires development of specific chemical probes, the alternative thermal proteome profiling (TPP) approach, which neither requires chemical modification of the active compounds nor covalent interaction with the target protein, is gaining popularity [Citation23]. It uses multiplexed quantitative proteomic MS to monitor ligand binding-induced changes in the melting profile of proteins in complex proteomic samples. In combination with TMT-10plex labeling the method has been recently employed by Corpaz-Lopez et al. to identify L. donovani NMT as the most significant target of their potent pyrazolyl sulfonamide antileishmanial compound [Citation24]. Despite the use of highly multiplexed isobaric labeling and high-resolution Orbitrap mass spectrometers, TPP studies, as of writing, report only MS2-based quantifications. It should also be noted that the TPP is limited to soluble proteins and not all protein-ligand interactions will produce a noticeable change in the melting temperature. Nevertheless, as a promising technology, further development in the TPP and its increased application in several laboratories could offer new insights into Leishmania spp. biology.

5. Concluding remarks

Proteomics is arguably the most appropriate method for unraveling the complex biology of Leishmania spp. We have discussed selected applications and the realistic possibilities of proteomic MS technology in the study of Leishmania parasites (). It is anticipated that further developments in proteomic MS technologies, particularly with improvements in detection sensitivity, improved sample preparation methods and the combination of proteomics with chemical approaches such as activity-based protein profiling and genomic and transcriptomic technologies, will shed more light on the biology of Leishmania spp. and the molecular mechanisms of pathogenesis.

Table 1. Select examples of application and possibilities of proteomic MS in Leishmania spp

6. Expert opinion

Genome editing technologies such as CRISPR/Cas9 and Di-Cre are now increasingly being developed and applied for probing the essentiality of select Leishmania genes [Citation25]. Similarly, the opposite gain-of-function approach using COSMID overexpression library generation and sequencing, termed Cos-Seq [Citation26], is also catching up, particularly for applications such as drug target profiling in Leishmania spp. However, the special dependency of protein-based post-transcriptional mechanisms of gene expression regulation in Leishmania spp. calls for integrated applications of these gene manipulation approaches with robust and sensitive proteomic methods for unraveling the unique eukaryotic biology of this evolutionarily distant parasite.

Declaration of interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Reviewer disclosures

Peer reviewers in this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

K. Kalesh and P.W. Denny are funded, in part, by UK Research and Innovation via the Global Challenges Research Fund under grant agreement ‘A Global Network for Neglected Tropical Diseases’ grant number MR/P027989/1.

References

  • Aebersold R, Agar JN, Amster IJ, et al. How many human proteoforms are there? Nat Chem Biol. 2018;14(3):206–214.
  • Santos R, Ursu O, Gaulton A, et al. A comprehensive map of molecular drug targets. Nat Rev Drug Discov. 2017;16(1):19–34.
  • Burza S, Croft SL, Boelaert M. Leishmaniasis. Lancet. 2018;392(10151):951–970.
  • Kramer S. Developmental regulation of gene expression in the absence of transcriptional control: the case of kinetoplastids. Mol Biochem Parasitol. 2012;181(2):61–72.
  • Parthasarathy A, Kalesh K. Defeating the tripanosomatid trio: proteomics of the protozoan parasites causing neglected tropical diseases. RSC Med Chem. 2020;11:625–645.
  • Kalesh K, Denny PW. A BONCAT-iTRAQ method enables temporally resolved quantitative profiling of newly synthesised proteins in Leishmania mexicana parasites during starvation. PLoS Negl Trop Dis. 2019;13(12):e0007651.
  • Savitski MM, Zinn N, Faelth-Savitski M, et al., Multiplexed proteome dynamics profiling reveals mechanisms controlling protein homeostasis. Cell. 2018;173(1):260–274.
  • Tsigankov P, Gherardini PF, Helmer-Citterich M, et al. Phosphoproteomic analysis of differentiating Leishmania parasites reveals a unique stage-specific phosphorylation motif. J Proteome Res. 2013;12(7):3405–3412.
  • Mule SN, Saad JS, Fernandes LR, et al. Protein glycosylation in Leishmania spp. Mol Omics. 2020;16(5):407–424.
  • Rosenzweig D, Smith D, Myler PJ, et al. Post-translational modification of cellular proteins during Leishmania donovani differentiation. Proteomics. 2008;8(9):1843–1850.
  • Burge RJ, Damianou A, Wilkinson AJ, et al. Leishmania differentiation requires ubiquitin conjugation mediated by a UBC2-UEV1 E2 complex. PLoS Pathog. 2020;16(10):e1008784.
  • Damianou A, Burge RJ, CMC C-P, et al. Essential roles for deubiquitination in Leishmania life cycle progression. PLoS Pathog. 2020;16(6):e1008455.
  • Mulvey CM, Breckels LM, Geladaki A, et al., Using hyperLOPIT to perform high-resolution mapping of the spatial proteome. Nat Protoc. 2017;12(6):1110–1135.
  • Barylyuk K, Koreny L, Ke H, et al. A comprehensive subcellular atlas of the Toxoplasma proteome via hyperLOPIT provides spatial context for protein functions. Cell Host Microbe. 2020;28(5):752–766.
  • Menezes JP, Almeida TF, Petersen AL, et al. Proteomic analysis reveals differentially expressed proteins in macrophages infected with Leishmania amazonensis or Leishmania major. Microbes Infect. 2013;15(8–9):579–591.
  • Singh AK, Pandey RK, Siqueira-Neto JL, et al. Proteomic-based approach to gain insight into reprogramming of THP-1 cells exposed to Leishmania donovani over an early temporal window. Infect Immun. 2015;83(5):1853–1868.
  • Negrao F, Giorgio S, Eberlin MN, et al., Comparative proteomic analysis of murine cutaneous lesions induced by Leishmania amazonensis or Leishmania major. ACS Infect Dis. 2019;5(8):1295–1305. .
  • da Silva Santos C, Attarha S, Saini RK, et al. Proteome profiling of human cutaneous leishmaniasis lesion. J Invest Dermatol. 2015;135(2):400–410.
  • Bag AK, Saha S, Sundar S, et al. Comparative proteomics and glycoproteomics of plasma proteins in Indian visceral leishmaniasis. Proteome Sci. 2014;12(1):48.
  • Marr AK, MacIsaac JL, Jiang R, et al. Leishmania donovani infection causes distinct epigenetic DNA methylation changes in host macrophages. PLoS Pathog. 2014;10(10):e1004419.
  • Wyllie S, Thomas M, Patterson S, et al. Cyclin-dependent kinase 12 is a drug target for visceral leishmaniasis. Nature. 2018;560(7717):192–197.
  • Wright MH, Paape D, Storck EM, et al. Global analysis of protein N-myristoylation and exploration of N-myristoyltransferase as a drug target in the neglected human pathogen Leishmania donovani. Chem Biol. 2015;22(3):342–354.
  • Franken H, Mathieson T, Childs D, et al. Thermal proteome profiling for unbiased identification of direct and indirect drug targets using multiplexed quantitative mass spectrometry. Nat Protoc. 2015;10(10):1567–1593.
  • Corpas-Lopez V, Moniz S, Thomas M, et al. Pharmacological validation of N-myristoyltransferase as a drug target in Leishmania donovani. ACS Infect Dis. 2019;5(1):111–122.
  • Jones NG, CMC C-P, Lima A, et al. Genetically validated drug targets in Leishmania: current knowledge and future prospects. ACS Infect Dis. 2018;4(4):467–477.
  • Gazanion E, Fernandez-Prada C, Papadopoulou B, et al., Cos-Seq for high-throughput identification of drug target and resistance mechanisms in the protozoan parasite Leishmania. Proc Natl Acad Sci U S A. 2016;113(21):E3012–21.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.