1
|
Mahanta PJ, Lhouvum K. Plasmodium falciparum proteases as new drug targets with special focus on metalloproteases. Mol Biochem Parasitol 2024; 258:111617. [PMID: 38554736 DOI: 10.1016/j.molbiopara.2024.111617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/15/2024] [Accepted: 03/10/2024] [Indexed: 04/02/2024]
Abstract
Malaria poses a significant global health threat particularly due to the prevalence of Plasmodium falciparum infection. With the emergence of parasite resistance to existing drugs including the recently discovered artemisinin, ongoing research seeks novel therapeutic avenues within the malaria parasite. Proteases are promising drug targets due to their essential roles in parasite biology, including hemoglobin digestion, merozoite invasion, and egress. While exploring the genomic landscape of Plasmodium falciparum, it has been revealed that there are 92 predicted proteases, with only approximately 14 of them having been characterized. These proteases are further distributed among 26 families grouped into five clans: aspartic proteases, cysteine proteases, metalloproteases, serine proteases, and threonine proteases. Focus on metalloprotease class shows further role in organelle processing for mitochondria and apicoplasts suggesting the potential of metalloproteases as viable drug targets. Holistic understanding of the parasite intricate life cycle and identification of potential drug targets are essential for developing effective therapeutic strategies against malaria and mitigating its devastating global impact.
Collapse
Affiliation(s)
| | - Kimjolly Lhouvum
- Department of Biotechnology, National Institute of Technology, Arunachal Pradesh, India.
| |
Collapse
|
2
|
Hollin T, Abel S, Banks C, Hristov B, Prudhomme J, Hales K, Florens L, Stafford Noble W, Le Roch KG. Proteome-Wide Identification of RNA-dependent proteins and an emerging role for RNAs in Plasmodium falciparum protein complexes. Nat Commun 2024; 15:1365. [PMID: 38355719 PMCID: PMC10866993 DOI: 10.1038/s41467-024-45519-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 01/26/2024] [Indexed: 02/16/2024] Open
Abstract
Ribonucleoprotein complexes are composed of RNA, RNA-dependent proteins (RDPs) and RNA-binding proteins (RBPs), and play fundamental roles in RNA regulation. However, in the human malaria parasite, Plasmodium falciparum, identification and characterization of these proteins are particularly limited. In this study, we use an unbiased proteome-wide approach, called R-DeeP, a method based on sucrose density gradient ultracentrifugation, to identify RDPs. Quantitative analysis by mass spectrometry identifies 898 RDPs, including 545 proteins not yet associated with RNA. Results are further validated using a combination of computational and molecular approaches. Overall, this method provides the first snapshot of the Plasmodium protein-protein interaction network in the presence and absence of RNA. R-DeeP also helps to reconstruct Plasmodium multiprotein complexes based on co-segregation and deciphers their RNA-dependence. One RDP candidate, PF3D7_0823200, is functionally characterized and validated as a true RBP. Using enhanced crosslinking and immunoprecipitation followed by high-throughput sequencing (eCLIP-seq), we demonstrate that this protein interacts with various Plasmodium non-coding transcripts, including the var genes and ap2 transcription factors.
Collapse
Affiliation(s)
- Thomas Hollin
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA, USA
| | - Steven Abel
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA, USA
| | - Charles Banks
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Borislav Hristov
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Jacques Prudhomme
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA, USA
| | - Kianna Hales
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | | | - William Stafford Noble
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | - Karine G Le Roch
- Department of Molecular, Cell and Systems Biology, University of California Riverside, Riverside, CA, USA.
| |
Collapse
|
3
|
Gonçalves AF, Lima-Pinheiro A, Teixeira M, Cassiano GC, Cravo P, Ferreira PE. Mutation in the 26S proteasome regulatory subunit rpn2 gene in Plasmodium falciparum confers resistance to artemisinin. Front Cell Infect Microbiol 2024; 14:1342856. [PMID: 38404287 PMCID: PMC10884193 DOI: 10.3389/fcimb.2024.1342856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 01/22/2024] [Indexed: 02/27/2024] Open
Abstract
Introduction Malaria parasites increasingly develop resistance to all drugs available in the market, hampering the goal of reducing malaria burden. Methods Herein, we evaluated the impact of a single-nucleotide variant, E738K, present in the 26S proteasome regulatory subunit rpn2 gene, identified in Plasmodium chabaudi resistant parasites. Plasmids carrying a functional rpn2 interspecies chimeric gene with 5' recombination region from P. falciparum and 3' from P. chabaudi were constructed and transfected into Dd2 P. falciparum parasites. Results and discussion The 738K variant parasite line presented increased parasite survival when subjected to dihydroartemisinin (DHA), as well as increased chymotrypsin-like activity and decreased accumulation of polyubiquitinated proteins. We thus conclude that the ubiquitin-proteasome pathway, including the 738K variant, play an important role in parasite response to DHA, being the first report of a mutation in a potential DHA drug target enhancing parasite survival and contributing to a significant advance in the understanding the biology of artemisinin resistance.
Collapse
Affiliation(s)
- Adriana F. Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute (ICVS)/ Biomaterials, Biodegradables and Biomimetics Research Group (3B's)-PT Government Associate Laboratory, Braga, Portugal
| | - Ana Lima-Pinheiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute (ICVS)/ Biomaterials, Biodegradables and Biomimetics Research Group (3B's)-PT Government Associate Laboratory, Braga, Portugal
| | - Miguel Teixeira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute (ICVS)/ Biomaterials, Biodegradables and Biomimetics Research Group (3B's)-PT Government Associate Laboratory, Braga, Portugal
- Department of Protection of Specific Crops, InnovPlantProtect Collaborative Laboratory, Elvas, Portugal
| | - Gustavo Capatti Cassiano
- Global Health and Tropical Medicine (GHTM), Associate Laboratory in Translation and Innovation Towards Global Health (LA-REAL), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Lisbon, Portugal
| | - Pedro Cravo
- Global Health and Tropical Medicine (GHTM), Associate Laboratory in Translation and Innovation Towards Global Health (LA-REAL), Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa (UNL), Lisbon, Portugal
| | - Pedro E. Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- Life and Health Sciences Research Institute (ICVS)/ Biomaterials, Biodegradables and Biomimetics Research Group (3B's)-PT Government Associate Laboratory, Braga, Portugal
| |
Collapse
|
4
|
Lucky AB, Wang C, Shakri AR, Kalamuddin M, Chim-Ong A, Li X, Miao J. Plasmodium falciparum GCN5 plays a key role in regulating artemisinin resistance-related stress responses. Antimicrob Agents Chemother 2023; 67:e0057723. [PMID: 37702516 PMCID: PMC10583690 DOI: 10.1128/aac.00577-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/24/2023] [Indexed: 09/14/2023] Open
Abstract
Plasmodium falciparum causes the most severe malaria and is exposed to various environmental and physiological stresses in the human host. Given that GCN5 plays a critical role in regulating stress responses in model organisms, we aimed to elucidate PfGCN5's function in stress responses in P. falciparum. The protein level of PfGCN5 was substantially induced under three stress conditions [heat shock, low glucose starvation, and dihydroartemisinin, the active metabolite of artemisinin (ART)]. With a TetR-DOZI conditional knockdown (KD) system, we successfully down-regulated PfGCN5 to ~50% and found that KD parasites became more sensitive to all three stress conditions. Transcriptomic analysis via RNA-seq identified ~1,000 up- and down-regulated genes in the wild-type (WT) and KD parasites under these stress conditions. Importantly, DHA induced transcriptional alteration of many genes involved in many aspects of stress responses, which were heavily shared among the altered genes under heat shock and low glucose conditions, including ART-resistance-related genes such as K13 and coronin. Based on the expression pattern between WT and KD parasites under three stress conditions, ~300-400 genes were identified to be involved in PfGCN5-dependent, general, and stress-condition-specific responses with high levels of overlaps among three stress conditions. Notably, using ring-stage survival assay, we found that KD or inhibition of PfGCN5 could sensitize the ART-resistant parasites to the DHA treatment. All these indicate that PfGCN5 is pivotal in regulating general and ART-resistance-related stress responses in malaria parasites, implicating PfGCN5 as a potential target for malaria intervention.
Collapse
Affiliation(s)
- Amuza Byaruhanga Lucky
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Chengqi Wang
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Ahmad Rushdi Shakri
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Mohammad Kalamuddin
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Anongruk Chim-Ong
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Xiaolian Li
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Jun Miao
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
5
|
Rashpa R, Klages N, Schvartz D, Pasquarello C, Brochet M. The Skp1-Cullin1-FBXO1 complex is a pleiotropic regulator required for the formation of gametes and motile forms in Plasmodium berghei. Nat Commun 2023; 14:1312. [PMID: 36898988 PMCID: PMC10006092 DOI: 10.1038/s41467-023-36999-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 02/27/2023] [Indexed: 03/12/2023] Open
Abstract
Malaria-causing parasites of the Plasmodium genus undergo multiple developmental phases in the human and the mosquito hosts, regulated by various post-translational modifications. While ubiquitination by multi-component E3 ligases is key to regulate a wide range of cellular processes in eukaryotes, little is known about its role in Plasmodium. Here we show that Plasmodium berghei expresses a conserved SKP1/Cullin1/FBXO1 (SCFFBXO1) complex showing tightly regulated expression and localisation across multiple developmental stages. It is key to cell division for nuclear segregation during schizogony and centrosome partitioning during microgametogenesis. It is additionally required for parasite-specific processes including gamete egress from the host erythrocyte, as well as integrity of the apical and the inner membrane complexes (IMC) in merozoite and ookinete, two structures essential for the dissemination of these motile stages. Ubiquitinomic surveys reveal a large set of proteins ubiquitinated in a FBXO1-dependent manner including proteins important for egress and IMC organisation. We additionally demonstrate an interplay between FBXO1-dependent ubiquitination and phosphorylation via calcium-dependent protein kinase 1. Altogether we show that Plasmodium SCFFBXO1 plays conserved roles in cell division and is also important for parasite-specific processes in the mammalian and mosquito hosts.
Collapse
Affiliation(s)
- Ravish Rashpa
- University of Geneva, Faculty of Medicine, Department of Microbiology and Molecular Medicine, Geneva, Switzerland.
| | - Natacha Klages
- University of Geneva, Faculty of Medicine, Department of Microbiology and Molecular Medicine, Geneva, Switzerland
| | - Domitille Schvartz
- University of Geneva, Faculty of Medicine, Proteomics Core Facility, Geneva, Switzerland
| | - Carla Pasquarello
- University of Geneva, Faculty of Medicine, Proteomics Core Facility, Geneva, Switzerland
| | - Mathieu Brochet
- University of Geneva, Faculty of Medicine, Department of Microbiology and Molecular Medicine, Geneva, Switzerland.
| |
Collapse
|
6
|
Zhang H, Ginn J, Zhan W, Leung A, Liu YJ, Toita A, Okamoto R, Wong TT, Imaeda T, Hara R, Michino M, Yukawa T, Chelebieva S, Tumwebaze PK, Vendome J, Beuming T, Sato K, Aso K, Rosenthal PJ, Cooper RA, Liverton N, Foley M, Meinke PT, Nathan CF, Kirkman LA, Lin G. Structure-Activity Relationship Studies of Antimalarial Plasmodium Proteasome Inhibitors─Part II. J Med Chem 2023; 66:1484-1508. [PMID: 36630286 PMCID: PMC10157299 DOI: 10.1021/acs.jmedchem.2c01651] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
With increasing reports of resistance to artemisinins and artemisinin-combination therapies, targeting the Plasmodium proteasome is a promising strategy for antimalarial development. We recently reported a highly selective Plasmodium falciparum proteasome inhibitor with anti-malarial activity in the humanized mouse model. To balance the permeability of the series of macrocycles with other drug-like properties, we conducted further structure-activity relationship studies on a biphenyl ether-tethered macrocyclic scaffold. Extensive SAR studies around the P1, P3, and P5 groups and peptide backbone identified compound TDI-8414. TDI-8414 showed nanomolar antiparasitic activity, no toxicity to HepG2 cells, high selectivity against the Plasmodium proteasome over the human constitutive proteasome and immunoproteasome, improved solubility and PAMPA permeability, and enhanced metabolic stability in microsomes and plasma of both humans and mice.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA
| | - John Ginn
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St., New York, NY 10065, USA
| | - Wenhu Zhan
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA
| | - Annie Leung
- Divison of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA
| | - Yi J. Liu
- Divison of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA
| | - Akinori Toita
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St., New York, NY 10065, USA
| | - Rei Okamoto
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St., New York, NY 10065, USA
| | - Tzu-Tshin Wong
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St., New York, NY 10065, USA
| | - Toshihiro Imaeda
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St., New York, NY 10065, USA
| | - Ryoma Hara
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St., New York, NY 10065, USA
| | - Mayako Michino
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St., New York, NY 10065, USA
| | - Takafumi Yukawa
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St., New York, NY 10065, USA
| | - Sevil Chelebieva
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, CA 94901, USA
| | | | | | | | - Kenjiro Sato
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St., New York, NY 10065, USA
| | - Kazuyoshi Aso
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St., New York, NY 10065, USA
| | - Philip J. Rosenthal
- Department of Medicine, University of California, San Francisco, CA 94143, USA
| | - Roland A. Cooper
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, CA 94901, USA
| | - Nigel Liverton
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St., New York, NY 10065, USA
| | - Michael Foley
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St., New York, NY 10065, USA
| | - Peter T. Meinke
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St., New York, NY 10065, USA
| | - Carl F. Nathan
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA
| | - Laura A. Kirkman
- Divison of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA
| | - Gang Lin
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, USA
| |
Collapse
|
7
|
Susceptibilities of Ugandan Plasmodium falciparum Isolates to Proteasome Inhibitors. Antimicrob Agents Chemother 2022; 66:e0081722. [PMID: 36094216 PMCID: PMC9578402 DOI: 10.1128/aac.00817-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The proteasome is a promising target for antimalarial chemotherapy. We assessed ex vivo susceptibilities of fresh Plasmodium falciparum isolates from eastern Uganda to seven proteasome inhibitors: two asparagine ethylenediamines, two macrocyclic peptides, and three peptide boronates; five had median IC50 values <100 nM. TDI8304, a macrocylic peptide lead compound with drug-like properties, had a median IC50 of 16 nM. Sequencing genes encoding the β2 and β5 catalytic proteasome subunits, the predicted targets of the inhibitors, and five additional proteasome subunits, identified two mutations in β2 (I204T, S214F), three mutations in β5 (V2I, A142S, D150E), and three mutations in other subunits. The β2 S214F mutation was associated with decreased susceptibility to two peptide boronates, with IC50s of 181 nM and 2635 nM against mutant versus 62 nM and 477 nM against wild type parasites for MMV1579506 and MMV1794229, respectively, although significance could not be formally assessed due to the small number of mutant parasites with available data. The other β2 and β5 mutations and mutations in other subunits were not associated with susceptibility to tested compounds. Against culture-adapted Ugandan isolates, two asparagine ethylenediamines and the peptide proteasome inhibitors WLW-vinyl sulfone and WLL-vinyl sulfone (which were not studied ex vivo) demonstrated low nM activity, without decreased activity against β2 S214F mutant parasites. Overall, proteasome inhibitors had potent activity against P. falciparum isolates circulating in Uganda, and genetic variation in proteasome targets was uncommon.
Collapse
|
8
|
Lai JW, Maah MJ, Sarip R, Lim YAL, Tim KL, Ng CH. Potency of copper(II) complexes towards drug-sensitive and -resistant Plasmodium falciparum: structure-activity relationship, ROS-generation and proteasome inhibition. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
9
|
Serrano-Aparicio N, Ferrer S, Świderek K. Covalent Inhibition of the Human 20S Proteasome with Homobelactosin C Inquired by QM/MM Studies. Pharmaceuticals (Basel) 2022; 15:ph15050531. [PMID: 35631358 PMCID: PMC9143130 DOI: 10.3390/ph15050531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 12/02/2022] Open
Abstract
20S proteasome is a main player in the protein degradation pathway in the cytosol, thus intervening in multiple pivotal cellular processes. Over the years the proteasome has emerged as a crucial target for the treatment of many diseases such as neurodegenerative diseases, cancer, autoimmune diseases, developmental disorders, cystic fibrosis, diabetes, cardiac diseases, atherosclerosis, and aging. In this work, the mechanism of proteasome covalent inhibition with bisbenzyl-protected homobelactosin C (hBelC) was explored using quantum mechanics/molecular mechanics (QM/MM) methods. Molecular dynamic simulations were used to describe key interactions established between the hBelC and its unique binding mode in the primed site of the β5 subunit. The free energy surfaces were computed to characterize the kinetics and thermodynamics of the inhibition process. This study revealed that although the final inhibition product for hBelC is formed according to the same molecular mechanism as one described for hSalA, the free energy profile of the reaction pathway differs significantly from the one previously reported for γ-lactam-β-lactone containing inhibitors in terms of the height of the activation barrier as well as the stabilization of the final product. Moreover, it was proved that high stabilization of the covalent adduct formed between β5-subunit and hBelC, together with the presence of aminocarbonyl side chain in the structure of the inhibitor which prevents the hydrolysis of the ester bond from taking place, determines its irreversible character.
Collapse
|
10
|
Miyasaka Y, Niwa S, Masuya T, Ishii R, Kobayashi M, Horio F, Ohno T. E3 ubiquitin ligase RNF123-deficient mice exhibit reduced parasitemia and mortality in rodent malaria (Plasmodium yoelii 17XL) infection. Parasitol Int 2022; 88:102542. [PMID: 35063657 DOI: 10.1016/j.parint.2022.102542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/22/2021] [Accepted: 01/12/2022] [Indexed: 11/19/2022]
Abstract
Increased levels of several human ubiquitin ligases, including ring finger protein 123 (RNF123), in red blood cells with Plasmodium falciparum infection, have been reported. RNF123 is an E3 ubiquitin ligase that is highly expressed in erythroid cells. However, the function of the RNF123 gene and the relationship between the RNF123 gene and malarial parasite has not been clarified in vivo. In this study, we generated RNF123-deficient mice using the CRISPR/Cas9 system, and analyzed malaria susceptibility and erythrocyte morphology. The levels of parasitemia 5 days post-infection and mortality 21 days post-infection with the lethal type of rodent malaria (Plasmodium yoelii 17XL) in RNF123-deficient mice was significantly lower than that in wild-type mice. In contrast, red blood cell morphology in RNF123-deficient mice was almost normal. These results suggest that erythrocytic RNF123 plays a role in susceptibility to rodent malaria infection, but does not play a role in erythrocyte morphology.
Collapse
Affiliation(s)
- Yuki Miyasaka
- Division of Experimental Animals, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | - Shota Niwa
- Division of Experimental Animals, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | - Tomomi Masuya
- Department of Applied Molecular Bioscience, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8601, Japan
| | - Reika Ishii
- Department of Applied Molecular Bioscience, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8601, Japan
| | - Misato Kobayashi
- Department of Applied Molecular Bioscience, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8601, Japan; Department of Nutritional Sciences, Nagoya University of Arts and Sciences, 57 Takenoyama, Iwasaki-cho, Nisshin, Aichi 470-0196, Japan
| | - Fumihiko Horio
- Department of Applied Molecular Bioscience, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8601, Japan; Department of Life Studies and Environmental Science, Nagoya Women's University, 3-40 Shioji-cho, Mizuho-ku, Nagoya, Aichi 467-8610, Japan
| | - Tamio Ohno
- Division of Experimental Animals, Graduate School of Medicine, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan.
| |
Collapse
|
11
|
Bharti H, Singal A, Saini M, Cheema PS, Raza M, Kundu S, Nag A. Repurposing the Pathogen Box compounds for identification of potent anti-malarials against blood stages of Plasmodium falciparum with PfUCHL3 inhibitory activity. Sci Rep 2022; 12:918. [PMID: 35042884 PMCID: PMC8766476 DOI: 10.1038/s41598-021-04619-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 12/22/2021] [Indexed: 11/08/2022] Open
Abstract
Malaria has endured as a global epidemic since ages and its eradication poses an immense challenge due to the complex life cycle of the causative pathogen and its tolerance to a myriad of therapeutics. PfUCHL3, a member of the ubiquitin C-terminal hydrolase (UCH) family of deubiquitinases (DUBs) is cardinal for parasite survival and emerges as a promising therapeutic target. In this quest, we employed a combination of computational and experimental approaches to identify PfUCHL3 inhibitors as novel anti-malarials. The Pathogen Box library was screened against the crystal structure of PfUCHL3 (PDB ID: 2WE6) and its human ortholog (PDB ID: 1XD3). Fifty molecules with better comparative score, bioavailability and druglikeliness were subjected to in-vitro enzyme inhibition assay and among them only two compounds effectively inhibited PfUCHL3 activity at micro molar concentrations. Both MMV676603 and MMV688704 exhibited anti-plasmodial activity by altering the parasite phenotype at late stages of the asexual life cycle and inducing the accumulation of polyubiquitinated substrates. In addition, both the compounds were non-toxic and portrayed high selectivity window for the parasite over mammalian cells. This is the first comprehensive study to demonstrate the anti-malarial efficacy of PfUCHL3 inhibitors and opens new avenues to exploit UCH family of DUBs as a promising target for the development of next generation anti-malaria therapy.
Collapse
Affiliation(s)
- Hina Bharti
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021, India
| | - Aakriti Singal
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021, India
| | - Manisha Saini
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021, India
| | - Pradeep Singh Cheema
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021, India
| | - Mohsin Raza
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021, India
| | - Suman Kundu
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021, India
| | - Alo Nag
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021, India.
| |
Collapse
|
12
|
Onchieku NM, Kumari S, Pandey R, Sharma V, Kumar M, Deshmukh A, Kaur I, Mohmmed A, Gupta D, Kiboi D, Gaur N, Malhotra P. Artemisinin Binds and Inhibits the Activity of Plasmodium falciparum Ddi1, a Retroviral Aspartyl Protease. Pathogens 2021; 10:pathogens10111465. [PMID: 34832620 PMCID: PMC8621276 DOI: 10.3390/pathogens10111465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 01/10/2023] Open
Abstract
Reduced sensitivity of the human malaria parasite, Plasmodium falciparum, to Artemisinin and its derivatives (ARTs) threatens the global efforts towards eliminating malaria. ARTs have been shown to cause ubiquitous cellular and genetic insults, which results in the activation of the unfolded protein response (UPR) pathways. The UPR restores protein homeostasis, which otherwise would be toxic to cellular survival. Here, we interrogated the role of DNA-damage inducible protein 1 (PfDdi1), a unique proteasome-interacting retropepsin in mediating the actions of the ARTs. We demonstrate that PfDdi1 is an active A2 family protease that hydrolyzes ubiquitinated proteasome substrates. Treatment of P. falciparum parasites with ARTs leads to the accumulation of ubiquitinated proteins in the parasites and blocks the destruction of ubiquitinated proteins by inhibiting the PfDdi1 protease activity. Besides, whereas the PfDdi1 is predominantly localized in the cytoplasm, exposure of the parasites to ARTs leads to DNA fragmentation and increased recruitment of the PfDdi1 into the nucleus. Furthermore, we show that Ddi1 knock-out Saccharomycescerevisiae cells are more susceptible to ARTs and the PfDdI1 protein robustly restores the corresponding functions in the knock-out cells. Together, these results show that ARTs act in multiple ways; by inducing DNA and protein damage and might be impairing the damage recovery by inhibiting the activity of PfDdi1, an essential ubiquitin-proteasome retropepsin.
Collapse
Affiliation(s)
- Noah Machuki Onchieku
- Malaria Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India or (N.M.O.); (V.S.); (A.D.); (I.K.)
| | - Sonam Kumari
- Yeast Biofuel Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India; (S.K.); (M.K.); (N.G.)
| | - Rajan Pandey
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India; (R.P.); (D.G.)
| | - Vaibhav Sharma
- Malaria Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India or (N.M.O.); (V.S.); (A.D.); (I.K.)
| | - Mohit Kumar
- Yeast Biofuel Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India; (S.K.); (M.K.); (N.G.)
| | - Arunaditya Deshmukh
- Malaria Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India or (N.M.O.); (V.S.); (A.D.); (I.K.)
| | - Inderjeet Kaur
- Malaria Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India or (N.M.O.); (V.S.); (A.D.); (I.K.)
| | - Asif Mohmmed
- Parasite Cell Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India;
| | - Dinesh Gupta
- Translational Bioinformatics Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India; (R.P.); (D.G.)
| | - Daniel Kiboi
- Department of Biochemistry, Jomo Kenyatta University of Agriculture and Technology, Nairobi P.O. Box 62000-00200, Kenya;
| | - Naseem Gaur
- Yeast Biofuel Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India; (S.K.); (M.K.); (N.G.)
| | - Pawan Malhotra
- Malaria Biology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi 110067, India or (N.M.O.); (V.S.); (A.D.); (I.K.)
- Correspondence: or
| |
Collapse
|
13
|
Maneekesorn S, Knuepfer E, Green JL, Prommana P, Uthaipibull C, Srichairatanakool S, Holder AA. Deletion of Plasmodium falciparum ubc13 increases parasite sensitivity to the mutagen, methyl methanesulfonate and dihydroartemisinin. Sci Rep 2021; 11:21791. [PMID: 34750454 PMCID: PMC8575778 DOI: 10.1038/s41598-021-01267-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/18/2021] [Indexed: 12/18/2022] Open
Abstract
The inducible Di-Cre system was used to delete the putative ubiquitin-conjugating enzyme 13 gene (ubc13) of Plasmodium falciparum to study its role in ubiquitylation and the functional consequence during the parasite asexual blood stage. Deletion resulted in a significant reduction of parasite growth in vitro, reduced ubiquitylation of the Lys63 residue of ubiquitin attached to protein substrates, and an increased sensitivity of the parasite to both the mutagen, methyl methanesulfonate and the antimalarial drug dihydroartemisinin (DHA), but not chloroquine. The parasite was also sensitive to the UBC13 inhibitor NSC697923. The data suggest that this gene does code for an ubiquitin conjugating enzyme responsible for K63 ubiquitylation, which is important in DNA repair pathways as was previously demonstrated in other organisms. The increased parasite sensitivity to DHA in the absence of ubc13 function indicates that DHA may act primarily through this pathway and that inhibitors of UBC13 may both enhance the efficacy of this antimalarial drug and directly inhibit parasite growth.
Collapse
Affiliation(s)
- Supawadee Maneekesorn
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Malaria Parasitology Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Ellen Knuepfer
- Malaria Parasitology Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Molecular and Cellular Parasitology Laboratory, Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hawkshead Lane, Hatfield, AL9 7TA, UK
| | - Judith L Green
- Malaria Parasitology Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Parichat Prommana
- Medical Molecular Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), 113 Thailand Science Park, Phahonyothin Road, Khlong Nueng, Khlong Luang, 12120, Pathum Thani, Thailand
| | - Chairat Uthaipibull
- Medical Molecular Biotechnology Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), 113 Thailand Science Park, Phahonyothin Road, Khlong Nueng, Khlong Luang, 12120, Pathum Thani, Thailand
- Thailand Center of Excellence for Life Sciences (TCELS), Phayathai, 10400, Bangkok, Thailand
| | - Somdet Srichairatanakool
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Anthony A Holder
- Malaria Parasitology Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| |
Collapse
|
14
|
Agamah FE, Damena D, Skelton M, Ghansah A, Mazandu GK, Chimusa ER. Network-driven analysis of human-Plasmodium falciparum interactome: processes for malaria drug discovery and extracting in silico targets. Malar J 2021; 20:421. [PMID: 34702263 PMCID: PMC8547565 DOI: 10.1186/s12936-021-03955-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/16/2021] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The emergence and spread of malaria drug resistance have resulted in the need to understand disease mechanisms and importantly identify essential targets and potential drug candidates. Malaria infection involves the complex interaction between the host and pathogen, thus, functional interactions between human and Plasmodium falciparum is essential to obtain a holistic view of the genetic architecture of malaria. Several functional interaction studies have extended the understanding of malaria disease and integrating such datasets would provide further insights towards understanding drug resistance and/or genetic resistance/susceptibility, disease pathogenesis, and drug discovery. METHODS This study curated and analysed data including pathogen and host selective genes, host and pathogen protein sequence data, protein-protein interaction datasets, and drug data from literature and databases to perform human-host and P. falciparum network-based analysis. An integrative computational framework is presented that was developed and found to be reasonably accurate based on various evaluations, applications, and experimental evidence of outputs produced, from data-driven analysis. RESULTS This approach revealed 8 hub protein targets essential for parasite and human host-directed malaria drug therapy. In a semantic similarity approach, 26 potential repurposable drugs involved in regulating host immune response to inflammatory-driven disorders and/or inhibiting residual malaria infection that can be appropriated for malaria treatment. Further analysis of host-pathogen network shortest paths enabled the prediction of immune-related biological processes and pathways subverted by P. falciparum to increase its within-host survival. CONCLUSIONS Host-pathogen network analysis reveals potential drug targets and biological processes and pathways subverted by P. falciparum to enhance its within malaria host survival. The results presented have implications for drug discovery and will inform experimental studies.
Collapse
Affiliation(s)
- Francis E Agamah
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Delesa Damena
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Michelle Skelton
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Anita Ghansah
- College of Health Sciences, Noguchi Memorial Institute for Medical Research, University of Ghana, P.O. Box LG 581, Legon, Ghana
| | - Gaston K Mazandu
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
- African Institute for Mathematical Sciences, 5-7 Melrose Road, Muizenberg, Cape Town, 7945, South Africa.
| | - Emile R Chimusa
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
15
|
Serrano-Aparicio N, Moliner V, Świderek K. On the Origin of the Different Reversible Characters of Salinosporamide A and Homosalinosporamide A in the Covalent Inhibition of the Human 20S Proteasome. ACS Catal 2021. [DOI: 10.1021/acscatal.1c02614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Natalia Serrano-Aparicio
- Biocomp Group, Institute of Advanced Materials (INAM), Universitat Jaume I, 12071 Castellón, Spain
| | - Vicent Moliner
- Biocomp Group, Institute of Advanced Materials (INAM), Universitat Jaume I, 12071 Castellón, Spain
| | - Katarzyna Świderek
- Biocomp Group, Institute of Advanced Materials (INAM), Universitat Jaume I, 12071 Castellón, Spain
| |
Collapse
|
16
|
Suratanee A, Buaboocha T, Plaimas K. Prediction of Human- Plasmodium vivax Protein Associations From Heterogeneous Network Structures Based on Machine-Learning Approach. Bioinform Biol Insights 2021; 15:11779322211013350. [PMID: 34188457 PMCID: PMC8212370 DOI: 10.1177/11779322211013350] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/04/2021] [Indexed: 11/24/2022] Open
Abstract
Malaria caused by Plasmodium vivax can lead to severe morbidity and death. In addition, resistance has been reported to existing drugs in treating this malaria. Therefore, the identification of new human proteins associated with malaria is urgently needed for the development of additional drugs. In this study, we established an analysis framework to predict human-P. vivax protein associations using network topological profiles from a heterogeneous network structure of human and P. vivax, machine-learning techniques and statistical analysis. Novel associations were predicted and ranked to determine the importance of human proteins associated with malaria. With the best-ranking score, 411 human proteins were identified as promising proteins. Their regulations and functions were statistically analyzed, which led to the identification of proteins involved in the regulation of membrane and vesicle formation, and proteasome complexes as potential targets for the treatment of P. vivax malaria. In conclusion, by integrating related data, our analysis was efficient in identifying potential targets providing an insight into human-parasite protein associations. Furthermore, generalizing this model could allow researchers to gain further insights into other diseases and enhance the field of biomedical science.
Collapse
Affiliation(s)
- Apichat Suratanee
- Department of Mathematics, Faculty of
Applied Science, King Mongkut’s University of Technology North Bangkok, Bangkok,
Thailand
| | - Teerapong Buaboocha
- Department of Biochemistry, Faculty of
Science, Chulalongkorn University, Bangkok, Thailand
- Omics Sciences and Bioinformatics
Center, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Kitiporn Plaimas
- Omics Sciences and Bioinformatics
Center, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Advanced Virtual and Intelligent
Computing (AVIC) Center, Department of Mathematics and Computer Science, Faculty of
Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
17
|
Sumam de Oliveira D, Kronenberger T, Palmisano G, Wrenger C, de Souza EE. Targeting SUMOylation in Plasmodium as a Potential Target for Malaria Therapy. Front Cell Infect Microbiol 2021; 11:685866. [PMID: 34178724 PMCID: PMC8224225 DOI: 10.3389/fcimb.2021.685866] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/10/2021] [Indexed: 11/13/2022] Open
Abstract
Malaria is a parasitic disease that represents a public health problem worldwide. Protozoans of the Plasmodium genus are responsible for causing malaria in humans. Plasmodium species have a complex life cycle that requires post-translational modifications (PTMs) to control cellular activities temporally and spatially and regulate the levels of critical proteins and cellular mechanisms for maintaining an efficient infection and immune evasion. SUMOylation is a PTM formed by the covalent linkage of a small ubiquitin-like modifier protein to the lysine residues on the protein substrate. This PTM is reversible and is triggered by the sequential action of three enzymes: E1-activating, E2-conjugating, and E3 ligase. On the other end, ubiquitin-like-protein-specific proteases in yeast and sentrin-specific proteases in mammals are responsible for processing SUMO peptides and for deconjugating SUMOylated moieties. Further studies are necessary to comprehend the molecular mechanisms and cellular functions of SUMO in Plasmodium. The emergence of drug-resistant malaria parasites prompts the discovery of new targets and antimalarial drugs with novel mechanisms of action. In this scenario, the conserved biological processes regulated by SUMOylation in the malaria parasites such as gene expression regulation, oxidative stress response, ubiquitylation, and proteasome pathways, suggest PfSUMO as a new potential drug target. This mini-review focuses on the current understanding of the mechanism of action of the PfSUMO during the coordinated multi-step life cycle of Plasmodium and discusses them as attractive new target proteins for the development of parasite-specific inhibitors and therapeutic intervention toward malaria disease.
Collapse
Affiliation(s)
- Daffiny Sumam de Oliveira
- Department of Parasitology, Institute of Biomedical Sciences at the University of São Paulo, São Paulo, Brazil
| | - Thales Kronenberger
- Department of Internal Medicine VIII, University Hospital Tübingen, Tübingen, Germany
| | - Giuseppe Palmisano
- Department of Parasitology, Institute of Biomedical Sciences at the University of São Paulo, São Paulo, Brazil
| | - Carsten Wrenger
- Department of Parasitology, Institute of Biomedical Sciences at the University of São Paulo, São Paulo, Brazil
| | - Edmarcia Elisa de Souza
- Department of Parasitology, Institute of Biomedical Sciences at the University of São Paulo, São Paulo, Brazil
| |
Collapse
|
18
|
Mukherjee P, Burgio G, Heitlinger E. Dual RNA Sequencing Meta-analysis in Plasmodium Infection Identifies Host-Parasite Interactions. mSystems 2021; 6:e00182-21. [PMID: 33879496 PMCID: PMC8546971 DOI: 10.1128/msystems.00182-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 03/04/2021] [Indexed: 11/20/2022] Open
Abstract
Dual RNA sequencing (RNA-Seq) is the simultaneous transcriptomic analysis of interacting symbionts, for example, in malaria. Potential cross-species interactions identified by correlated gene expression might highlight interlinked signaling, metabolic, or gene regulatory pathways in addition to physically interacting proteins. Often, malaria studies address one of the interacting organisms-host or parasite-rendering the other "contamination." Here we perform a meta-analysis using such studies for cross-species expression analysis. We screened experiments for gene expression from host and Plasmodium. Out of 171 studies in Homo sapiens, Macaca mulatta, and Mus musculus, we identified 63 potential studies containing host and parasite data. While 16 studies (1,950 samples) explicitly performed dual RNA-Seq, 47 (1,398 samples) originally focused on one organism. We found 915 experimental replicates from 20 blood studies to be suitable for coexpression analysis and used orthologs for meta-analysis across different host-parasite systems. Centrality metrics from the derived gene expression networks correlated with gene essentiality in the parasites. We found indications of host immune response to elements of the Plasmodium protein degradation system, an antimalarial drug target. We identified well-studied immune responses in the host with our coexpression networks, as our approach recovers known broad processes interlinked between hosts and parasites in addition to individual host and parasite protein associations. The set of core interactions represents commonalities between human malaria and its model systems for prioritization in laboratory experiments. Our approach might also allow insights into the transferability of model systems for different pathways in malaria studies.IMPORTANCE Malaria still causes about 400,000 deaths a year and is one of the most studied infectious diseases. The disease is studied in mice and monkeys as lab models to derive potential therapeutic intervention in human malaria. Interactions between Plasmodium spp. and its hosts are either conserved across different host-parasite systems or idiosyncratic to those systems. Here we use correlation of gene expression from different RNA-Seq studies to infer common host-parasite interactions across human, mouse, and monkey studies. First, we find a set of very conserved interactors, worth further scrutiny in focused laboratory experiments. Second, this work might help assess to which extent experiments and knowledge on different pathways can be transferred from models to humans for potential therapy.
Collapse
Affiliation(s)
- Parnika Mukherjee
- Department of Molecular Parasitology, Humboldt University, Berlin, Germany
- Research Group Ecology and Evolution of Molecular Parasite-Host Interactions, Leibniz-Institute for Zoo and Wildlife Research (IZW), Berlin, Germany
- Department of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Gaétan Burgio
- Department of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Emanuel Heitlinger
- Department of Molecular Parasitology, Humboldt University, Berlin, Germany
- Research Group Ecology and Evolution of Molecular Parasite-Host Interactions, Leibniz-Institute for Zoo and Wildlife Research (IZW), Berlin, Germany
| |
Collapse
|
19
|
Serrano-Aparicio N, Moliner V, Świderek K. Nature of Irreversible Inhibition of Human 20S Proteasome by Salinosporamide A. The Critical Role of Lys–Asp Dyad Revealed from Electrostatic Effects Analysis. ACS Catal 2021. [DOI: 10.1021/acscatal.0c05313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
| | - Vicent Moliner
- Departament de Química Física i Analítica, Universitat Jaume I, 12071 Castellón, Spain
| | - Katarzyna Świderek
- Departament de Química Física i Analítica, Universitat Jaume I, 12071 Castellón, Spain
| |
Collapse
|
20
|
20S proteasomes secreted by the malaria parasite promote its growth. Nat Commun 2021; 12:1172. [PMID: 33608523 PMCID: PMC7895969 DOI: 10.1038/s41467-021-21344-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/20/2021] [Indexed: 12/16/2022] Open
Abstract
Mature red blood cells (RBCs) lack internal organelles and canonical defense mechanisms, making them both a fascinating host cell, in general, and an intriguing choice for the deadly malaria parasite Plasmodium falciparum (Pf), in particular. Pf, while growing inside its natural host, the human RBC, secretes multipurpose extracellular vesicles (EVs), yet their influence on this essential host cell remains unknown. Here we demonstrate that Pf parasites, cultured in fresh human donor blood, secrete within such EVs assembled and functional 20S proteasome complexes (EV-20S). The EV-20S proteasomes modulate the mechanical properties of naïve human RBCs by remodeling their cytoskeletal network. Furthermore, we identify four degradation targets of the secreted 20S proteasome, the phosphorylated cytoskeletal proteins β-adducin, ankyrin-1, dematin and Epb4.1. Overall, our findings reveal a previously unknown 20S proteasome secretion mechanism employed by the human malaria parasite, which primes RBCs for parasite invasion by altering membrane stiffness, to facilitate malaria parasite growth. Plasmodium falciparum secretes extracellular vesicles (EVs) while growing inside red blood cells (RBCs). Here the authors show that these EVs contain assembled and functional 20S proteasome complexes that remodel the cytoskeleton of naïve human RBCs, priming the RBCs for parasite invasion.
Collapse
|
21
|
Simwela NV, Hughes KR, Rennie MT, Barrett MP, Waters AP. Mammalian Deubiquitinating Enzyme Inhibitors Display in Vitro and in Vivo Activity against Malaria Parasites and Potentiate Artemisinin Action. ACS Infect Dis 2021; 7:333-346. [PMID: 33400499 DOI: 10.1021/acsinfecdis.0c00580] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The ubiquitin proteasome system (UPS) is an emerging drug target in malaria due to its essential role in the parasite's life cycle stages as well its contribution to resistance to artemisinins. Polymorphisms in the Kelch13 gene of Plasmodium falciparum are primary markers of artemisinin resistance and among other things are phenotypically characterized by an overactive UPS. Inhibitors targeting the proteasome, critical components of the UPS, display activity in malaria parasites and synergize artemisinin action. Here we report the activity of small molecule inhibitors targeting mammalian deubiquitinating enzymes, DUBs (upstream UPS components), in malaria parasites. We show that generic DUB inhibitors can block intraerythrocytic development of malaria parasites in vitro and possess antiparasitic activity in vivo and can be used in combination with additive to synergistic effect. We also show that inhibition of these upstream components of the UPS can potentiate the activity of artemisinin in vitro as well as in vivo to the extent that artemisinin resistance can be overcome. Combinations of DUB inhibitors anticipated to target different DUB activities and downstream proteasome inhibitors are even more effective at improving the potency of artemisinins than either inhibitors alone, providing proof that targeting multiple UPS activities simultaneously could be an attractive approach to overcoming artemisinin resistance. These data further validate the parasite UPS as a target to both enhance artemisinin action and potentially overcome resistance. Lastly, we confirm that DUB inhibitors can be developed into in vivo antimalarial drugs with promise for activity against all of human malaria and could thus further exploit their current pursuit as anticancer agents in rapid drug repurposing programs.
Collapse
Affiliation(s)
- Nelson V. Simwela
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, Scotland G12 8TA, United Kingdom
| | - Katie R. Hughes
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, Scotland G12 8TA, United Kingdom
| | - Michael T. Rennie
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, Scotland G12 8TA, United Kingdom
| | - Michael P. Barrett
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, Scotland G12 8TA, United Kingdom
| | - Andrew P. Waters
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, Scotland G12 8TA, United Kingdom
| |
Collapse
|
22
|
Lu KY, Pasaje CFA, Srivastava T, Loiselle DR, Niles JC, Derbyshire E. Phosphatidylinositol 3-phosphate and Hsp70 protect Plasmodium falciparum from heat-induced cell death. eLife 2020; 9:e56773. [PMID: 32975513 PMCID: PMC7518890 DOI: 10.7554/elife.56773] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 09/14/2020] [Indexed: 12/12/2022] Open
Abstract
Phosphatidylinositol 3-phosphate (PI(3)P) levels in Plasmodium falciparum correlate with tolerance to cellular stresses caused by artemisinin and environmental factors. However, PI(3)P function during the Plasmodium stress response was unknown. Here, we used PI3K inhibitors and antimalarial agents to examine the importance of PI(3)P under thermal conditions recapitulating malarial fever. Live cell microscopy using chemical and genetic reporters revealed that PI(3)P stabilizes the digestive vacuole (DV) under heat stress. We demonstrate that heat-induced DV destabilization in PI(3)P-deficient P. falciparum precedes cell death and is reversible after withdrawal of the stress condition and the PI3K inhibitor. A chemoproteomic approach identified PfHsp70-1 as a PI(3)P-binding protein. An Hsp70 inhibitor and knockdown of PfHsp70-1 phenocopy PI(3)P-deficient parasites under heat shock. Furthermore, PfHsp70-1 downregulation hypersensitizes parasites to heat shock and PI3K inhibitors. Our findings underscore a mechanistic link between PI(3)P and PfHsp70-1 and present a novel PI(3)P function in DV stabilization during heat stress.
Collapse
Affiliation(s)
- Kuan-Yi Lu
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke UniversityDurhamUnited States
- Department of Chemistry, Duke UniversityDurhamUnited States
| | | | | | - David R Loiselle
- Department of Pharmacology and Cancer Biology, School of Medicine, Duke UniversityDurhamUnited States
| | - Jacquin C Niles
- Department of Biological Engineering, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Emily Derbyshire
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke UniversityDurhamUnited States
- Department of Chemistry, Duke UniversityDurhamUnited States
| |
Collapse
|
23
|
Moyo P, Mugumbate G, Eloff JN, Louw AI, Maharaj VJ, Birkholtz LM. Natural Products: A Potential Source of Malaria Transmission Blocking Drugs? Pharmaceuticals (Basel) 2020; 13:E251. [PMID: 32957668 PMCID: PMC7558993 DOI: 10.3390/ph13090251] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 12/17/2022] Open
Abstract
The ability to block human-to-mosquito and mosquito-to-human transmission of Plasmodium parasites is fundamental to accomplish the ambitious goal of malaria elimination. The WHO currently recommends only primaquine as a transmission-blocking drug but its use is severely restricted by toxicity in some populations. New, safe and clinically effective transmission-blocking drugs therefore need to be discovered. While natural products have been extensively investigated for the development of chemotherapeutic antimalarial agents, their potential use as transmission-blocking drugs is comparatively poorly explored. Here, we provide a comprehensive summary of the activities of natural products (and their derivatives) of plant and microbial origins against sexual stages of Plasmodium parasites and the Anopheles mosquito vector. We identify the prevailing challenges and opportunities and suggest how these can be mitigated and/or exploited in an endeavor to expedite transmission-blocking drug discovery efforts from natural products.
Collapse
Affiliation(s)
- Phanankosi Moyo
- Malaria Parasite Molecular Laboratory, Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, Faculty of Natural and Agricultural Sciences, University of Pretoria, Private Bag x20, Hatfield, 0028 Pretoria, South Africa;
| | - Grace Mugumbate
- Department of Chemistry, School of Natural Sciences and Mathematics, Chinhoyi University of Technology, Private Bag, 7724 Chinhoyi, Zimbabwe;
| | - Jacobus N. Eloff
- Phytomedicine Programme, Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Private Bag x04, Onderstepoort 0110 Pretoria, South Africa;
| | - Abraham I. Louw
- Malaria Parasite Molecular Laboratory, Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, Faculty of Natural and Agricultural Sciences, University of Pretoria, Private Bag x20, Hatfield, 0028 Pretoria, South Africa;
| | - Vinesh J. Maharaj
- Department of Chemistry, Faculty of Natural and Agricultural Sciences, University of Pretoria, Private Bag x20, Hatfield, 0028 Pretoria, South Africa;
| | - Lyn-Marié Birkholtz
- Malaria Parasite Molecular Laboratory, Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, Faculty of Natural and Agricultural Sciences, University of Pretoria, Private Bag x20, Hatfield, 0028 Pretoria, South Africa;
| |
Collapse
|
24
|
Basova S, Wilke N, Koch JC, Prokop A, Berkessel A, Pradel G, Ngwa CJ. Organoarsenic Compounds with In Vitro Activity against the Malaria Parasite Plasmodium falciparum. Biomedicines 2020; 8:biomedicines8080260. [PMID: 32748808 PMCID: PMC7459655 DOI: 10.3390/biomedicines8080260] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/24/2020] [Accepted: 07/29/2020] [Indexed: 01/16/2023] Open
Abstract
The rapid development of parasite drug resistance as well as the lack of medications targeting both the asexual and the sexual blood stages of the malaria parasite necessitate the search for novel antimalarial compounds. Eleven organoarsenic compounds were synthesized and tested for their effect on the asexual blood stages and sexual transmission stages of the malaria parasite Plasmodium falciparum using in vitro assays. The inhibitory potential of the compounds on blood stage viability was tested on the chloroquine (CQ)-sensitive 3D7 and the CQ-resistant Dd2 strain using the Malstat assay. The most effective compounds were subsequently investigated for their effect on impairing gametocyte development and gametogenesis, using the gametocyte-producing NF54 strain in respective cell-based assays. Their potential toxicity was investigated on leukemia cell line Nalm-6 and non-infected erythrocytes. Five out of the 11 compounds showed antiplasmodial activities against 3D7, with half-maximal inhibitory concentration (IC50) values ranging between 1.52 and 8.64 µM. Three of the compounds also acted against Dd2, with the most active compound As-8 exhibiting an IC50 of 0.35 µM. The five compounds also showed significant inhibitory effects on the parasite sexual stages at both IC50 and IC90 concentrations with As-8 displaying the best gametocytocidal activity. No hemolytic and cytotoxic effect was observed for any of the compounds. The organoarsenic compound As-8 may represent a good lead for the design of novel organoarsenic drugs with combined antimalarial and transmission blocking activities.
Collapse
Affiliation(s)
- Sofia Basova
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany; (S.B.); (G.P.)
| | - Nathalie Wilke
- Department of Paediatric Oncology, Children’s Hospital Cologne, Amsterdamer Straße 59, 50735 Cologne, Germany; (N.W.); (A.P.)
| | - Jan Christoph Koch
- Department of Chemistry, Organic Chemistry, University of Cologne, Greinstraße 4, 50939 Cologne, Germany; (J.C.K.); (A.B.)
| | - Aram Prokop
- Department of Paediatric Oncology, Children’s Hospital Cologne, Amsterdamer Straße 59, 50735 Cologne, Germany; (N.W.); (A.P.)
- Department of Paediatric Oncology, Helios Hospital Schwerin, Wismarsche Strasse 393-397, 19049 Schwerin, Germany
| | - Albrecht Berkessel
- Department of Chemistry, Organic Chemistry, University of Cologne, Greinstraße 4, 50939 Cologne, Germany; (J.C.K.); (A.B.)
| | - Gabriele Pradel
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany; (S.B.); (G.P.)
| | - Che Julius Ngwa
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany; (S.B.); (G.P.)
- Correspondence:
| |
Collapse
|
25
|
Green JL, Wu Y, Encheva V, Lasonder E, Prommaban A, Kunzelmann S, Christodoulou E, Grainger M, Truongvan N, Bothe S, Sharma V, Song W, Pinzuti I, Uthaipibull C, Srichairatanakool S, Birault V, Langsley G, Schindelin H, Stieglitz B, Snijders AP, Holder AA. Ubiquitin activation is essential for schizont maturation in Plasmodium falciparum blood-stage development. PLoS Pathog 2020; 16:e1008640. [PMID: 32569299 PMCID: PMC7332102 DOI: 10.1371/journal.ppat.1008640] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/02/2020] [Accepted: 05/17/2020] [Indexed: 11/19/2022] Open
Abstract
Ubiquitylation is a common post translational modification of eukaryotic proteins and in the human malaria parasite, Plasmodium falciparum (Pf) overall ubiquitylation increases in the transition from intracellular schizont to extracellular merozoite stages in the asexual blood stage cycle. Here, we identify specific ubiquitylation sites of protein substrates in three intraerythrocytic parasite stages and extracellular merozoites; a total of 1464 sites in 546 proteins were identified (data available via ProteomeXchange with identifier PXD014998). 469 ubiquitylated proteins were identified in merozoites compared with only 160 in the preceding intracellular schizont stage, suggesting a large increase in protein ubiquitylation associated with merozoite maturation. Following merozoite invasion of erythrocytes, few ubiquitylated proteins were detected in the first intracellular ring stage but as parasites matured through trophozoite to schizont stages the apparent extent of ubiquitylation increased. We identified commonly used ubiquitylation motifs and groups of ubiquitylated proteins in specific areas of cellular function, for example merozoite pellicle proteins involved in erythrocyte invasion, exported proteins, and histones. To investigate the importance of ubiquitylation we screened ubiquitin pathway inhibitors in a parasite growth assay and identified the ubiquitin activating enzyme (UBA1 or E1) inhibitor MLN7243 (TAK-243) to be particularly effective. This small molecule was shown to be a potent inhibitor of recombinant PfUBA1, and a structural homology model of MLN7243 bound to the parasite enzyme highlights avenues for the development of P. falciparum specific inhibitors. We created a genetically modified parasite with a rapamycin-inducible functional deletion of uba1; addition of either MLN7243 or rapamycin to the recombinant parasite line resulted in the same phenotype, with parasite development blocked at the schizont stage. Nuclear division and formation of intracellular structures was interrupted. These results indicate that the intracellular target of MLN7243 is UBA1, and this activity is essential for the final differentiation of schizonts to merozoites.
Collapse
Affiliation(s)
- Judith L. Green
- Malaria Parasitology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Yang Wu
- Malaria Parasitology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Vesela Encheva
- Mass Spectrometry Proteomics, The Francis Crick Institute, London, United Kingdom
| | - Edwin Lasonder
- School of Biomedical Science, University of Plymouth, Plymouth, United Kingdom
| | - Adchara Prommaban
- Malaria Parasitology Laboratory, The Francis Crick Institute, London, United Kingdom
- Department of Biochemistry, Chiang Mai University, Chiang Mai, Thailand
| | - Simone Kunzelmann
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Evangelos Christodoulou
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Munira Grainger
- Malaria Parasitology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Ngoc Truongvan
- Rudolf Virchow Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany
| | - Sebastian Bothe
- Department of Chemistry and Pharmacy, University of Würzburg, Würzburg, Germany
| | - Vikram Sharma
- School of Biomedical Science, University of Plymouth, Plymouth, United Kingdom
| | - Wei Song
- School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom
| | - Irene Pinzuti
- School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom
| | - Chairat Uthaipibull
- National Center for Genetic Engineering and Biotechnology, Khlong Luang, Thailand
| | | | | | - Gordon Langsley
- Laboratoire de Biologie Cellulaire Comparative des Apicomplexes, Institut Cochin, Université Paris Descartes, Paris, France
| | - Hermann Schindelin
- Rudolf Virchow Center for Experimental Biomedicine, Universität Würzburg, Würzburg, Germany
| | - Benjamin Stieglitz
- School of Biological and Chemical Sciences, Queen Mary University of London, London, United Kingdom
| | | | - Anthony A. Holder
- Malaria Parasitology Laboratory, The Francis Crick Institute, London, United Kingdom
- * E-mail:
| |
Collapse
|
26
|
Giannangelo C, Siddiqui G, De Paoli A, Anderson BM, Edgington-Mitchell LE, Charman SA, Creek DJ. System-wide biochemical analysis reveals ozonide antimalarials initially act by disrupting Plasmodium falciparum haemoglobin digestion. PLoS Pathog 2020; 16:e1008485. [PMID: 32589689 PMCID: PMC7347234 DOI: 10.1371/journal.ppat.1008485] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/09/2020] [Accepted: 05/13/2020] [Indexed: 01/23/2023] Open
Abstract
Ozonide antimalarials, OZ277 (arterolane) and OZ439 (artefenomel), are synthetic peroxide-based antimalarials with potent activity against the deadliest malaria parasite, Plasmodium falciparum. Here we used a "multi-omics" workflow, in combination with activity-based protein profiling (ABPP), to demonstrate that peroxide antimalarials initially target the haemoglobin (Hb) digestion pathway to kill malaria parasites. Time-dependent metabolomic profiling of ozonide-treated P. falciparum infected red blood cells revealed a rapid depletion of short Hb-derived peptides followed by subsequent alterations in lipid and nucleotide metabolism, while untargeted peptidomics showed accumulation of longer Hb-derived peptides. Quantitative proteomics and ABPP assays demonstrated that Hb-digesting proteases were increased in abundance and activity following treatment, respectively. Ozonide-induced depletion of short Hb-derived peptides was less extensive in a drug-treated K13-mutant artemisinin resistant parasite line (Cam3.IIR539T) than in the drug-treated isogenic sensitive strain (Cam3.IIrev), further confirming the association between ozonide activity and Hb catabolism. To demonstrate that compromised Hb catabolism may be a primary mechanism involved in ozonide antimalarial activity, we showed that parasites forced to rely solely on Hb digestion for amino acids became hypersensitive to short ozonide exposures. Quantitative proteomics analysis also revealed parasite proteins involved in translation and the ubiquitin-proteasome system were enriched following drug treatment, suggestive of the parasite engaging a stress response to mitigate ozonide-induced damage. Taken together, these data point to a mechanism of action involving initial impairment of Hb catabolism, and indicate that the parasite regulates protein turnover to manage ozonide-induced damage.
Collapse
Affiliation(s)
- Carlo Giannangelo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Ghizal Siddiqui
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Amanda De Paoli
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Bethany M. Anderson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Laura E. Edgington-Mitchell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Department of Maxillofacial Surgery, College of Dentistry, New York University, New York, New York, United States of America
| | - Susan A. Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Darren J. Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
27
|
Plasmodium chaperonin TRiC/CCT identified as a target of the antihistamine clemastine using parallel chemoproteomic strategy. Proc Natl Acad Sci U S A 2020; 117:5810-5817. [PMID: 32127489 DOI: 10.1073/pnas.1913525117] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The antihistamine clemastine inhibits multiple stages of the Plasmodium parasite that causes malaria, but the molecular targets responsible for its parasite inhibition were unknown. Here, we applied parallel chemoproteomic platforms to discover the mechanism of action of clemastine and identify that clemastine binds to the Plasmodium falciparum TCP-1 ring complex or chaperonin containing TCP-1 (TRiC/CCT), an essential heterooligomeric complex required for de novo cytoskeletal protein folding. Clemastine destabilized all eight P. falciparum TRiC subunits based on thermal proteome profiling (TPP). Further analysis using stability of proteins from rates of oxidation (SPROX) revealed a clemastine-induced thermodynamic stabilization of the Plasmodium TRiC delta subunit, suggesting an interaction with this protein subunit. We demonstrate that clemastine reduces levels of the major TRiC substrate tubulin in P. falciparum parasites. In addition, clemastine treatment leads to disorientation of Plasmodium mitotic spindles during the asexual reproduction and results in aberrant tubulin morphology suggesting protein aggregation. This clemastine-induced disruption of TRiC function is not observed in human host cells, demonstrating a species selectivity required for targeting an intracellular human pathogen. Our findings encourage larger efforts to apply chemoproteomic methods to assist in target identification of antimalarial drugs and highlight the potential to selectively target Plasmodium TRiC-mediated protein folding for malaria intervention.
Collapse
|
28
|
Proteasome Inhibitors: Harnessing Proteostasis to Combat Disease. Molecules 2020; 25:molecules25030671. [PMID: 32033280 PMCID: PMC7037493 DOI: 10.3390/molecules25030671] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/25/2020] [Accepted: 01/28/2020] [Indexed: 02/07/2023] Open
Abstract
The proteasome is the central component of the main cellular protein degradation pathway. During the past four decades, the critical function of the proteasome in numerous physiological processes has been revealed, and proteasome activity has been linked to various human diseases. The proteasome prevents the accumulation of misfolded proteins, controls the cell cycle, and regulates the immune response, to name a few important roles for this macromolecular "machine." As a therapeutic target, proteasome inhibitors have been approved for the treatment of multiple myeloma and mantle cell lymphoma. However, inability to sufficiently inhibit proteasome activity at tolerated doses has hampered efforts to expand the scope of proteasome inhibitor-based therapies. With emerging new modalities in myeloma, it might seem challenging to develop additional proteasome-based therapies. However, the constant development of new applications for proteasome inhibitors and deeper insights into the intricacies of protein homeostasis suggest that proteasome inhibitors might have novel therapeutic applications. Herein, we summarize the latest advances in proteasome inhibitor development and discuss the future of proteasome inhibitors and other proteasome-based therapies in combating human diseases.
Collapse
|
29
|
Sindhe KMV, Wu W, Legac J, Zhang YK, Easom EE, Cooper RA, Plattner JJ, Freund YR, DeRisi JL, Rosenthal PJ. Plasmodium falciparum Resistance to a Lead Benzoxaborole Due to Blocked Compound Activation and Altered Ubiquitination or Sumoylation. mBio 2020; 11:e02640-19. [PMID: 31992618 PMCID: PMC6989105 DOI: 10.1128/mbio.02640-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 12/05/2019] [Indexed: 02/04/2023] Open
Abstract
New antimalarial drugs are needed. The benzoxaborole AN13762 showed excellent activity against cultured Plasmodium falciparum, against fresh Ugandan P. falciparum isolates, and in murine malaria models. To gain mechanistic insights, we selected in vitro for P. falciparum isolates resistant to AN13762. In all of 11 independent selections with 100 to 200 nM AN13762, the 50% inhibitory concentration (IC50) increased from 18-118 nM to 180-890 nM, and whole-genome sequencing of resistant parasites demonstrated mutations in prodrug activation and resistance esterase (PfPARE). The introduction of PfPARE mutations led to a similar level of resistance, and recombinant PfPARE hydrolyzed AN13762 to the benzoxaborole AN10248, which has activity similar to that of AN13762 but for which selection of resistance was not readily achieved. Parasites further selected with micromolar concentrations of AN13762 developed higher-level resistance (IC50, 1.9 to 5.0 μM), and sequencing revealed additional mutations in any of 5 genes, 4 of which were associated with ubiquitination/sumoylation enzyme cascades; the introduction of one of these mutations, in SUMO-activating enzyme subunit 2, led to a similar level of resistance. The other gene mutated in highly resistant parasites encodes the P. falciparum cleavage and specificity factor homolog PfCPSF3, previously identified as the antimalarial target of another benzoxaborole. Parasites selected for resistance to AN13762 were cross-resistant with a close analog, AN13956, but not with standard antimalarials, AN10248, or other benzoxaboroles known to have different P. falciparum targets. Thus, AN13762 appears to have a novel mechanism of antimalarial action and multiple mechanisms of resistance, including loss of function of PfPARE preventing activation to AN10248, followed by alterations in ubiquitination/sumoylation pathways or PfCPSF3.IMPORTANCE Benzoxaboroles are under study as potential new drugs to treat malaria. One benzoxaborole, AN13762, has potent activity and promising features, but its mechanisms of action and resistance are unknown. To gain insights into these mechanisms, we cultured malaria parasites with nonlethal concentrations of AN13762 and generated parasites with varied levels of resistance. Parasites with low-level resistance had mutations in PfPARE, which processes AN13762 into an active metabolite; PfPARE mutations prevented this processing. Parasites with high-level resistance had mutations in any of a number of enzymes, mostly those involved in stress responses. Parasites selected for AN13762 resistance were not resistant to other antimalarials, suggesting novel mechanisms of action and resistance for AN13762, a valuable feature for a new class of antimalarial drugs.
Collapse
Affiliation(s)
- Kirthana M V Sindhe
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Wesley Wu
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | - Jenny Legac
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | | | - Eric E Easom
- Anacor Pharmaceuticals, Inc., Palo Alto, California, USA
| | - Roland A Cooper
- Dominican University of California, San Rafael, California, USA
| | | | | | - Joseph L DeRisi
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | - Philip J Rosenthal
- Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
30
|
Tibon NS, Ng CH, Cheong SL. Current progress in antimalarial pharmacotherapy and multi-target drug discovery. Eur J Med Chem 2019; 188:111983. [PMID: 31911292 DOI: 10.1016/j.ejmech.2019.111983] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 12/09/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022]
Abstract
Discovery and development of antimalarial drugs have long been dominated by single-target therapy. Continuous effort has been made to explore and identify different targets in malaria parasite crucial for the malaria treatment. The single-target drug therapy was initially successful, but it was later supplanted by combination therapy with multiple drugs to overcome drug resistance. Emergence of resistant strains even against the combination therapy has warranted a review of current antimalarial pharmacotherapy. This has led to the development of the new concept of covalent biotherapy, in which two or more pharmacophores are chemically bound to produce hybrid antimalarial drugs with multi-target functionalities. Herein, the review initially details the current pharmacotherapy for malaria as well as the conventional and novel targets of importance identified in the malaria parasite. Then, the rationale of multi-targeted therapy for malaria, approaches taken to develop the multi-target antimalarial hybrids, and the examples of hybrid molecules are comprehensively enumerated and discussed.
Collapse
Affiliation(s)
- Natasha Stella Tibon
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Chew Hee Ng
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| | - Siew Lee Cheong
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
31
|
Yadav DK, Kumar S, Teli MK, Yadav R, Chaudhary S. Molecular Targets for Malarial Chemotherapy: A Review. Curr Top Med Chem 2019; 19:861-873. [DOI: 10.2174/1568026619666190603080000] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 02/08/2019] [Accepted: 02/08/2019] [Indexed: 11/22/2022]
Abstract
The malaria parasite resistance to the existing drugs is a serious problem to the currently used
antimalarials and, thus, highlights the urgent need to develop new and effective anti-malarial molecules.
This could be achieved either by the identification of the new drugs for the validated targets or by further
refining/improving the existing antimalarials; or by combining previously effective agents with
new/existing drugs to have a synergistic effect that counters parasite resistance; or by identifying novel
targets for the malarial chemotherapy. In this review article, a comprehensive collection of some of the
novel molecular targets has been enlisted for the antimalarial drugs. The targets which could be deliberated
for developing new anti-malarial drugs could be: membrane biosynthesis, mitochondrial system,
apicoplasts, parasite transporters, shikimate pathway, hematin crystals, parasite proteases, glycolysis,
isoprenoid synthesis, cell cycle control/cycline dependent kinase, redox system, nucleic acid metabolism,
methionine cycle and the polyamines, folate metabolism, the helicases, erythrocyte G-protein, and
farnesyl transferases. Modern genomic tools approaches such as structural biology and combinatorial
chemistry, novel targets could be identified followed by drug development for drug resistant strains providing
wide ranges of novel targets in the development of new therapy. The new approaches and targets
mentioned in the manuscript provide a basis for the development of new unique strategies for antimalarial
therapy with limited off-target effects in the near future.
Collapse
Affiliation(s)
- Dharmendra K. Yadav
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro, 191, Yeonsu-gu, Incheon 406-799, South Korea
| | - Surendra Kumar
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro, 191, Yeonsu-gu, Incheon 406-799, South Korea
| | - Mahesh K. Teli
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro, 191, Yeonsu-gu, Incheon 406-799, South Korea
| | - Ravikant Yadav
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, Malaviya National Institute of Technology, Jawaharlal Nehru Marg, Jaipur-302017, India
| | - Sandeep Chaudhary
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, Malaviya National Institute of Technology, Jawaharlal Nehru Marg, Jaipur-302017, India
| |
Collapse
|
32
|
Effect of Metabolite Extract of Streptomyces hygroscopicus subsp. hygroscopicus on Plasmodium falciparum 3D7 in Vitro. IRANIAN JOURNAL OF PARASITOLOGY 2019; 14:444-452. [PMID: 31673263 PMCID: PMC6815862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Malaria eradication has been complicated by the repeated emergence of antimalarial drug resistances. We aimed to determine whether a metabolite extract of Streptomyces hygrocopicus subsp. hygroscopicus could decrease the viability of Plasmodium falciparum 3D7 in vitro. METHODS S. hygroscopicus subsp. hygroscopicus isolates were inoculated and fermented on the ISP4 medium. The fermented S. hygroscopicus was mixed with ethylacetate 1:5 (v/v), and the solvent phase was evaporated. Several concentrations of isolated extract was added to the P. falciparum 3D7 culture containing trophozoite and schizont stages in 24 wells plates when the degree of parasite-infected erythrocytes reached 5%, then incubated for 8 hours. DNA parasite density was measured using flow cytometry, parasite degree and morphology were observed under microscopic by Giemsa-stained smears. RESULTS The metabolite extract affected the morphology of almost all of parasite asexual stages. Schizonts and trophozoites failed to grow and appeared damaged with pycnotic cores and loss of cytoplasmic content. At 8 hours there was a significant decrease in DNA parasite density in culture exposed to 2.6 mg/ml and 13 mg/ml (P = 0.002; P = 0.024) of the extract. The degree of parasite-infected erythrocytes was decreased from the beginning of exposure (0.02 mg/ml of the extract). There was a significant inverse correlation between the concentration of extract and the degree of parasite-infected erythrocytes as well as the density of DNA parasite (r = -0.772, P = 0.000; r =-0.753; P =0.000). CONCLUSION Metabolite extract of S. hygroscopicus subsp. hygroscopicus causes morphological damage, decreases the degree of parasite-infected erythrocytes and the DNA density of P. falciparum 3D7 in vitro.
Collapse
|
33
|
Tripeptide analogues of MG132 as protease inhibitors. Bioorg Med Chem 2018; 27:436-441. [PMID: 30581047 DOI: 10.1016/j.bmc.2018.12.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 12/17/2022]
Abstract
The 26S proteasome and calpain are linked to a number of important human diseases. Here, we report a series of analogues of the prototypical tripeptide aldehyde inhibitor MG132 that show a unique combination of high activity and selectivity for calpains over proteasome. Tripeptide aldehydes (1-3) with an aromatic P3 substituent show enhanced activity and selectivity against ovine calpain 2 relative to chymotrypsin-like activity of proteasome. Docking studies reveal the key contacts between inhibitors and calpain to confirm the importance of the S3 pocket with respect to selectivity between calpains 1 and 2 and the proteasome.
Collapse
|
34
|
Krishnan KM, Williamson KC. The proteasome as a target to combat malaria: hits and misses. Transl Res 2018; 198:40-47. [PMID: 30009761 PMCID: PMC6422032 DOI: 10.1016/j.trsl.2018.04.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/26/2018] [Accepted: 04/30/2018] [Indexed: 01/25/2023]
Abstract
The proteasome plays a vital role throughout the life cycle as Plasmodium parasites quickly adapt to a new host and undergo a series of morphologic changes during asexual replication and sexual differentiation. Plasmodium carries 3 different types of protease complexes: typical eukaryotic proteasome (26S) that resides in the cytoplasm and the nucleus, a prokaryotic proteasome homolog ClpQ that resides in the mitochondria, and a caseinolytic protease complex ClpP that resides in the apicoplast. In silico prediction in conjunction with immunoprecipitation analysis of ubiquitin conjugates have suggested that over half of the Plasmodium falciparum proteome during asexual reproduction are potential targets for ubiquitination. The marked potency of multiple classes of proteasome inhibitors against all stages of the life cycle, synergy with the current frontline antimalarial, artemisinin, and recent advances identifying differences between Plasmodium and human proteasomes strongly support further drug development efforts.
Collapse
Affiliation(s)
| | - Kim C Williamson
- Uniformed Services University of the Health Sciences, Bethesda, Maryland.
| |
Collapse
|
35
|
Escotte-Binet S, Huguenin A, Aubert D, Martin AP, Kaltenbach M, Florent I, Villena I. Metallopeptidases of Toxoplasma gondii: in silico identification and gene expression. ACTA ACUST UNITED AC 2018; 25:26. [PMID: 29737275 PMCID: PMC5939537 DOI: 10.1051/parasite/2018025] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 04/16/2018] [Indexed: 12/17/2022]
Abstract
Metallopeptidases are a family of proteins with domains that remain highly conserved throughout evolution. These hydrolases require divalent metal cation(s) to activate the water molecule in order to carry out their catalytic action on peptide bonds by nucleophilic attack. Metallopeptidases from parasitic protozoa, including Toxoplasma, are investigated because of their crucial role in parasite biology. In the present study, we screened the T. gondii database using PFAM motifs specific for metallopeptidases in association with the MEROPS peptidase Database (release 10.0). In all, 49 genes encoding proteins with metallopeptidase signatures were identified in the Toxoplasma genome. An Interpro Search enabled us to uncover their domain/motif organization, and orthologs with the highest similarity by BLAST were used for annotation. These 49 Toxoplasma metallopeptidases clustered into 15 families described in the MEROPS database. Experimental expression analysis of their genes in the tachyzoite stage revealed transcription for all genes studied. Further research on the role of these peptidases should increase our knowledge of basic Toxoplasma biology and provide opportunities to identify novel therapeutic targets. This type of study would also open a path towards the comparative biology of apicomplexans.
Collapse
Affiliation(s)
- Sandie Escotte-Binet
- EA 7510, ESCAPE, Laboratory of Parasitology-Mycology, University of Reims Champagne-Ardenne, 51100 Reims, France - Laboratory of Parasitology-Mycology, Toxoplasmosis National Reference Center, Toxoplasma Biological Resource Center, Maison Blanche Hospital, 51100 Reims, France
| | - Antoine Huguenin
- EA 7510, ESCAPE, Laboratory of Parasitology-Mycology, University of Reims Champagne-Ardenne, 51100 Reims, France - Laboratory of Parasitology-Mycology, Toxoplasmosis National Reference Center, Toxoplasma Biological Resource Center, Maison Blanche Hospital, 51100 Reims, France
| | - Dominique Aubert
- EA 7510, ESCAPE, Laboratory of Parasitology-Mycology, University of Reims Champagne-Ardenne, 51100 Reims, France - Laboratory of Parasitology-Mycology, Toxoplasmosis National Reference Center, Toxoplasma Biological Resource Center, Maison Blanche Hospital, 51100 Reims, France
| | - Anne-Pascaline Martin
- EA 7510, ESCAPE, Laboratory of Parasitology-Mycology, University of Reims Champagne-Ardenne, 51100 Reims, France
| | - Matthieu Kaltenbach
- EA 7510, ESCAPE, Laboratory of Parasitology-Mycology, University of Reims Champagne-Ardenne, 51100 Reims, France
| | - Isabelle Florent
- UMR7245 CNRS-MNHN, National Museum of Natural History, Department Adaptations of the Living, 75005 Paris, France
| | - Isabelle Villena
- EA 7510, ESCAPE, Laboratory of Parasitology-Mycology, University of Reims Champagne-Ardenne, 51100 Reims, France - Laboratory of Parasitology-Mycology, Toxoplasmosis National Reference Center, Toxoplasma Biological Resource Center, Maison Blanche Hospital, 51100 Reims, France
| |
Collapse
|
36
|
Cromm PM, Crews CM. The Proteasome in Modern Drug Discovery: Second Life of a Highly Valuable Drug Target. ACS CENTRAL SCIENCE 2017; 3:830-838. [PMID: 28852696 PMCID: PMC5571462 DOI: 10.1021/acscentsci.7b00252] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Indexed: 06/07/2023]
Abstract
As the central figure of the cellular protein degradation machinery, the proteasome is critical for cell survival. Having been extensively targeted for inhibition, the constitutive proteasome has proven its role as a highly valuable drug target. However, recent advances in the protein homeostasis field suggest that additional chapters can be added to this successful story. For example, selective immunoproteasome inhibition promises high clinical efficacy for autoimmune disorders and inflammation, and proteasome inhibitors might serve as novel therapeutics for malaria or other microorganisms. Furthermore, utilizing the destructive force of the proteasome for selective degradation of essential drivers of human disorders has opened up a new and exciting area of drug discovery. Thus, the field of proteasome drug discovery still holds exciting questions to be answered and does not simply end with inhibiting the constitutive proteasome.
Collapse
Affiliation(s)
- Philipp M. Cromm
- Department
of Molecular, Cellular & Developmental Biology, Yale University, New Haven, Connecticut 06511, United States
| | - Craig M. Crews
- Department
of Molecular, Cellular & Developmental Biology, Yale University, New Haven, Connecticut 06511, United States
- Department
of Chemistry, Yale University, New Haven, Connecticut 06511, United States
- Department
of Pharmacology, Yale University, New Haven, Connecticut 06511, United States
| |
Collapse
|
37
|
Ngwa CJ, Kiesow MJ, Papst O, Orchard LM, Filarsky M, Rosinski AN, Voss TS, Llinás M, Pradel G. Transcriptional Profiling Defines Histone Acetylation as a Regulator of Gene Expression during Human-to-Mosquito Transmission of the Malaria Parasite Plasmodium falciparum. Front Cell Infect Microbiol 2017; 7:320. [PMID: 28791254 PMCID: PMC5522858 DOI: 10.3389/fcimb.2017.00320] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/28/2017] [Indexed: 12/16/2022] Open
Abstract
Transmission of the malaria parasite Plasmodium falciparum from the human to the mosquito is mediated by the intraerythrocytic gametocytes, which, once taken up during a blood meal, become activated to initiate sexual reproduction. Because gametocytes are the only parasite stages able to establish an infection in the mosquito, they are crucial for spreading the tropical disease. During gametocyte maturation, different repertoires of genes are switched on and off in a well-coordinated sequence, pointing to regulatory mechanisms of gene expression. While epigenetic gene control has been studied during erythrocytic schizogony of P. falciparum, little is known about this process during human-to-mosquito transmission of the parasite. To unveil the potential role of histone acetylation during gene expression in gametocytes, we carried out a microarray-based transcriptome analysis on gametocytes treated with the histone deacetylase inhibitor trichostatin A (TSA). TSA-treatment impaired gametocyte maturation and lead to histone hyper-acetylation in these stages. Comparative transcriptomics identified 294 transcripts, which were more than 2-fold up-regulated during gametocytogenesis following TSA-treatment. In activated gametocytes, which were less sensitive to TSA, the transcript levels of 48 genes were increased. TSA-treatment further led to repression of ~145 genes in immature and mature gametocytes and 7 genes in activated gametocytes. Up-regulated genes are mainly associated with functions in invasion, cytoadherence, and protein export, while down-regulated genes could particularly be assigned to transcription and translation. Chromatin immunoprecipitation demonstrated a link between gene activation and histone acetylation for selected genes. Among the genes up-regulated in TSA-treated mature gametocytes was a gene encoding the ring finger (RING)-domain protein PfRNF1, a putative E3 ligase of the ubiquitin-mediated signaling pathway. Immunochemistry demonstrated PfRNF1 expression mainly in the sexual stages of P. falciparum with peak expression in stage II gametocytes, where the protein localized to the nucleus and cytoplasm. Pfrnf1 promoter and coding regions associated with acetylated histones, and TSA-treatment resulted in increased PfRNF1 levels. Our combined data point to an essential role of histone acetylation for gene regulation in gametocytes, which can be exploited for malaria transmission-blocking interventions.
Collapse
Affiliation(s)
- Che J Ngwa
- Division of Cellular and Applied Infection Biology, RWTH Aachen UniversityAachen, Germany
| | - Meike J Kiesow
- Division of Cellular and Applied Infection Biology, RWTH Aachen UniversityAachen, Germany
| | - Olga Papst
- Division of Cellular and Applied Infection Biology, RWTH Aachen UniversityAachen, Germany
| | - Lindsey M Orchard
- Department of Biochemistry and Molecular Biology, The Pennsylvania State UniversityUniversity Park, PA, United States
| | - Michael Filarsky
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health InstituteBasel, Switzerland
| | - Alina N Rosinski
- Division of Cellular and Applied Infection Biology, RWTH Aachen UniversityAachen, Germany
| | - Till S Voss
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health InstituteBasel, Switzerland
| | - Manuel Llinás
- Department of Biochemistry and Molecular Biology, The Pennsylvania State UniversityUniversity Park, PA, United States.,Department of Chemistry and Huck Center for Malaria Research, The Pennsylvania State UniversityUniversity Park, PA, United States
| | - Gabriele Pradel
- Division of Cellular and Applied Infection Biology, RWTH Aachen UniversityAachen, Germany
| |
Collapse
|
38
|
Protein Degradation Systems as Antimalarial Therapeutic Targets. Trends Parasitol 2017; 33:731-743. [PMID: 28688800 DOI: 10.1016/j.pt.2017.05.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 12/11/2022]
Abstract
Artemisinin (ART)-based combination therapies are the most efficacious treatment of uncomplicated Plasmodium falciparum malaria. Alarmingly, P. falciparum strains have acquired resistance to ART across much of Southeast Asia. ART creates widespread protein and lipid damage inside intraerythrocytic parasites, necessitating macromolecule degradation. The proteasome is the main engine of Plasmodium protein degradation. Indeed, proteasome inhibition and ART have shown synergy in ART-resistant parasites. Moreover, ubiquitin modification is associated with altered parasite susceptibility to multiple antimalarials. Targeting the ubiquitin-proteasome system (UPS), therefore, is an attractive avenue to combat drug resistance. Here, we review recent advances leading to specific targeting of the Plasmodium proteasome. We also highlight the potential for targeting other nonproteasomal protein degradation systems as an additional strategy to disrupt protein homeostasis.
Collapse
|
39
|
González-López L, Carballar-Lejarazú R, Arrevillaga Boni G, Cortés-Martínez L, Cázares-Raga FE, Trujillo-Ocampo A, Rodríguez MH, James AA, Hernández-Hernández FDLC. Lys48 ubiquitination during the intraerythrocytic cycle of the rodent malaria parasite, Plasmodium chabaudi. PLoS One 2017; 12:e0176533. [PMID: 28604779 PMCID: PMC5467854 DOI: 10.1371/journal.pone.0176533] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 04/12/2017] [Indexed: 12/26/2022] Open
Abstract
Ubiquitination tags proteins for different functions within the cell. One of the most abundant and studied ubiquitin modification is the Lys48 polyubiquitin chain that modifies proteins for their destruction by proteasome. In Plasmodium is proposed that post-translational regulation is fundamental for parasite development during its complex life-cycle; thus, the objective of this work was to analyze the ubiquitination during Plasmodium chabaudi intraerythrocytic stages. Ubiquitinated proteins were detected during intraerythrocytic stages of Plasmodium chabaudi by immunofluorescent microscopy, bidimensional electrophoresis (2-DE) combined with immunoblotting and mass spectrometry. All the studied stages presented protein ubiquitination and Lys48 polyubiquitination with more abundance during the schizont stage. Three ubiquitinated proteins were identified for rings, five for trophozoites and twenty for schizonts. Only proteins detected with a specific anti- Lys48 polyubiquitin antibody were selected for Mass Spectrometry analysis and two of these identified proteins were selected in order to detect the specific amino acid residues where ubiquitin is placed. Ubiquitinated proteins during the ring and trophozoite stages were related with the invasion process and in schizont proteins were related with nucleic acid metabolism, glycolysis and protein biosynthesis. Most of the ubiquitin detection was during the schizont stage and the Lys48 polyubiquitination during this stage was related to proteins that are expected to be abundant during the trophozoite stage. The evidence that these Lys48 polyubiquitinated proteins are tagged for destruction by the proteasome complex suggests that this type of post-translational modification is important in the regulation of protein abundance during the life-cycle and may also contribute to the parasite cell-cycle progression.
Collapse
Affiliation(s)
- Lorena González-López
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), México, D.F., México
| | - Rebeca Carballar-Lejarazú
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), México, D.F., México
- Departments of Molecular Biology & Biochemistry and Microbiology & Molecular Genetics, University of California, Irvine, California, United States of America
| | - Gerardo Arrevillaga Boni
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), México, D.F., México
| | - Leticia Cortés-Martínez
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), México, D.F., México
| | - Febe Elena Cázares-Raga
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), México, D.F., México
| | - Abel Trujillo-Ocampo
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), México, D.F., México
| | - Mario H. Rodríguez
- Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Morelos, México
| | - Anthony A. James
- Departments of Molecular Biology & Biochemistry and Microbiology & Molecular Genetics, University of California, Irvine, California, United States of America
| | - Fidel de la Cruz Hernández-Hernández
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV-IPN), México, D.F., México
- * E-mail:
| |
Collapse
|
40
|
Devender N, Gunjan S, Tripathi R, Tripathi RP. Synthesis and antiplasmodial activity of novel indoleamide derivatives bearing sulfonamide and triazole pharmacophores. Eur J Med Chem 2017; 131:171-184. [PMID: 28319782 DOI: 10.1016/j.ejmech.2017.03.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 12/19/2022]
Abstract
Due to the recent reports of growing parasite resistance to artemisinins and other antimalarial drugs, development of new antimalarial chemotypes is an urgent priority. Here in, we report a novel series of adamantyl/cycloheptyl indoleamide derivatives bearing sulfonamide and triazole pharmacophores adopting different chemical modifications and evaluated them for antiplasmodial activity in vitro. Among all the indoleamides, compounds 22, 24, 26 and 30 with sulfonamide pharmacophore showed promising activity with IC50 of 1.87, 1.93, 2.00, 2.17 μM against CQ sensitive Pf3D7 strain and 1.69, 2.12, 1.60, 2.19 μM against CQ resistant PfK1 strain, respectively.
Collapse
Affiliation(s)
- N Devender
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Sarika Gunjan
- Parasitology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110001, India
| | - Renu Tripathi
- Parasitology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110001, India.
| | - Rama Pati Tripathi
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110001, India.
| |
Collapse
|
41
|
Bibo-Verdugo B, Jiang Z, Caffrey CR, O'Donoghue AJ. Targeting proteasomes in infectious organisms to combat disease. FEBS J 2017; 284:1503-1517. [PMID: 28122162 DOI: 10.1111/febs.14029] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/21/2016] [Accepted: 01/23/2017] [Indexed: 01/04/2023]
Abstract
Proteasomes are multisubunit, energy-dependent, proteolytic complexes that play an essential role in intracellular protein turnover. They are present in eukaryotes, archaea, and in some actinobacteria species. Inhibition of proteasome activity has emerged as a powerful strategy for anticancer therapy and three drugs have been approved for treatment of multiple myeloma. These compounds react covalently with a threonine residue located in the active site of a proteasome subunit to block protein degradation. Proteasomes in pathogenic organisms such as Mycobacterium tuberculosis and Plasmodium falciparum also have a nucleophilic threonine residue in the proteasome active site and are therefore sensitive to these anticancer drugs. This review summarizes efforts to validate the proteasome in pathogenic organisms as a therapeutic target. We describe several strategies that have been used to develop inhibitors with increased potency and selectivity for the pathogen proteasome relative to the human proteasome. In addition, we highlight a cell-based chemical screening approach that identified a potent, allosteric inhibitor of proteasomes found in Leishmania and Trypanosoma species. Finally, we discuss the development of proteasome inhibitors as anti-infective agents.
Collapse
Affiliation(s)
- Betsaida Bibo-Verdugo
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.,Center for Discovery and Innovation in Parasitic Diseases, University of California San Diego, La Jolla, CA, USA
| | - Zhenze Jiang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.,Chemistry & Biochemistry Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Conor R Caffrey
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.,Center for Discovery and Innovation in Parasitic Diseases, University of California San Diego, La Jolla, CA, USA
| | - Anthony J O'Donoghue
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.,Center for Discovery and Innovation in Parasitic Diseases, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
42
|
Tools for attenuation of gene expression in malaria parasites. Int J Parasitol 2017; 47:385-398. [PMID: 28153780 DOI: 10.1016/j.ijpara.2016.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/15/2016] [Accepted: 11/28/2016] [Indexed: 12/30/2022]
Abstract
An understanding of the biology of Plasmodium parasites, which are the causative agents of the disease malaria, requires study of gene function. Various reverse genetic tools have been described for determining gene function. These tools can be broadly grouped as trans- and cis-acting. Trans-acting tools control gene functions through synthetic nucleic acid probe molecules matching the sequence of the gene of interest. Once delivered to the parasite, the probe engages with the mRNA of the target gene and attenuates its function. Cis-acting tools control gene function through elements introduced into the gene of interest by DNA transfection. The expression of the modified gene can be controlled using external agents, typically small molecule ligands. In this review, we discuss the strengths and weaknesses of these tools to guide researchers in selecting the appropriate tool for studies of gene function, and for guiding future refinements of these tools.
Collapse
|
43
|
Ray A, Sarkar S. The proteasome of the differently-diverged eukaryote Giardia lamblia and its role in remodeling of the microtubule-based cytoskeleton. Crit Rev Microbiol 2016; 43:481-492. [PMID: 28033730 DOI: 10.1080/1040841x.2016.1262814] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Atrayee Ray
- Department of Biochemistry, Bose Institute, Kolkata, West Bengal, India
| | - Srimonti Sarkar
- Department of Biochemistry, Bose Institute, Kolkata, West Bengal, India
| |
Collapse
|
44
|
Low ML, Chan CW, Ng PY, Ooi IH, Maah MJ, Chye SM, Tan KW, Ng SW, Ng CH. Ternary and binary copper(II) complexes: synthesis, characterization, ROS-inductive, proteasome inhibitory, and anticancer properties. J COORD CHEM 2016. [DOI: 10.1080/00958972.2016.1260711] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- May Lee Low
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Cheang Wei Chan
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
- Department of Chemistry, Faculty of Science, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Pei Ying Ng
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Ing Hong Ooi
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Mohd Jamil Maah
- Department of Chemistry, Faculty of Science, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Soi Moi Chye
- Department of Human Biology, School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Kong Wai Tan
- Department of Chemistry, Faculty of Science, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Seik Weng Ng
- Department of Chemistry, Faculty of Science, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Chew Hee Ng
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
45
|
Structure and substrate fingerprint of aminopeptidase P from Plasmodium falciparum. Biochem J 2016; 473:3189-204. [DOI: 10.1042/bcj20160550] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 07/26/2016] [Indexed: 12/18/2022]
Abstract
Malaria is one of the world's most prevalent parasitic diseases, with over 200 million cases annually. Alarmingly, the spread of drug-resistant parasites threatens the effectiveness of current antimalarials and has made the development of novel therapeutic strategies a global health priority. Malaria parasites have a complicated lifecycle, involving an asymptomatic ‘liver stage’ and a symptomatic ‘blood stage’. During the blood stage, the parasites utilise a proteolytic cascade to digest host hemoglobin, which produces free amino acids absolutely necessary for parasite growth and reproduction. The enzymes required for hemoglobin digestion are therefore attractive therapeutic targets. The final step of the cascade is catalyzed by several metalloaminopeptidases, including aminopeptidase P (APP). We developed a novel platform to examine the substrate fingerprint of APP from Plasmodium falciparum (PfAPP) and to show that it can catalyze the removal of any residue immediately prior to a proline. Further, we have determined the crystal structure of PfAPP and present the first examination of the 3D structure of this essential malarial enzyme. Together, these analyses provide insights into potential mechanisms of inhibition that could be used to develop novel antimalarial therapeutics.
Collapse
|
46
|
Kaiser G, De Niz M, Zuber B, Burda PC, Kornmann B, Heussler VT, Stanway RR. High resolution microscopy reveals an unusual architecture of the Plasmodium berghei endoplasmic reticulum. Mol Microbiol 2016; 102:775-791. [PMID: 27566438 DOI: 10.1111/mmi.13490] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2016] [Indexed: 02/04/2023]
Abstract
To fuel the tremendously fast replication of Plasmodium liver stage parasites, the endoplasmic reticulum (ER) must play a critical role as a major site of protein and lipid biosynthesis. In this study, we analysed the parasite's ER morphology and function. Previous studies exploring the parasite ER have mainly focused on the blood stage. Visualizing the Plasmodium berghei ER during liver stage development, we found that the ER forms an interconnected network throughout the parasite with perinuclear and peripheral localizations. Surprisingly, we observed that the ER additionally generates huge accumulations. Using stimulated emission depletion microscopy and serial block-face scanning electron microscopy, we defined ER accumulations as intricate dense networks of ER tubules. We provide evidence that these accumulations are functional subdivisions of the parasite ER, presumably generated in response to elevated demands of the parasite, potentially consistent with ER stress. Compared to higher eukaryotes, Plasmodium parasites have a fundamentally reduced unfolded protein response machinery for reacting to ER stress. Accordingly, parasite development is greatly impaired when ER stress is applied. As parasites appear to be more sensitive to ER stress than are host cells, induction of ER stress could potentially be used for interference with parasite development.
Collapse
Affiliation(s)
- Gesine Kaiser
- Institute of Cell Biology, University of Bern, Baltzerstr. 4, 3012 Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Baltzerstrasse 2, 3012, Bern, Switzerland
| | - Mariana De Niz
- Institute of Cell Biology, University of Bern, Baltzerstr. 4, 3012 Bern, Switzerland.,Wellcome Trust Center for Molecular Parasitology, G12 8TA, Glasgow, UK
| | - Benoît Zuber
- Institute of Anatomy, University of Bern, Baltzerstrasse 2, 3012, Bern, Switzerland
| | - Paul-Christian Burda
- Institute of Cell Biology, University of Bern, Baltzerstr. 4, 3012 Bern, Switzerland
| | - Benoît Kornmann
- Institute of Biochemistry, ETH Zürich, Otto-Stern-Weg 3, 8093, Zürich, Switzerland
| | - Volker T Heussler
- Institute of Cell Biology, University of Bern, Baltzerstr. 4, 3012 Bern, Switzerland
| | - Rebecca R Stanway
- Institute of Cell Biology, University of Bern, Baltzerstr. 4, 3012 Bern, Switzerland
| |
Collapse
|
47
|
Structural characterization, ROS-inductive and proteasome inhibitory properties of ternary and binary copper(II) complexes of N2- and N2O2-ligands. Inorganica Chim Acta 2016. [DOI: 10.1016/j.ica.2016.06.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
48
|
Mata-Cantero L, Azkargorta M, Aillet F, Xolalpa W, LaFuente MJ, Elortza F, Carvalho AS, Martin-Plaza J, Matthiesen R, Rodriguez MS. New insights into host-parasite ubiquitin proteome dynamics in P. falciparum infected red blood cells using a TUBEs-MS approach. J Proteomics 2016; 139:45-59. [PMID: 26972027 DOI: 10.1016/j.jprot.2016.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 02/11/2016] [Accepted: 03/02/2016] [Indexed: 02/06/2023]
Abstract
UNLABELLED Malaria, caused by Plasmodium falciparum (P. falciparum), ranks as one of the most baleful infectious diseases worldwide. New antimalarial treatments are needed to face existing or emerging drug resistant strains. Protein degradation appears to play a significant role during the asexual intraerythrocytic developmental cycle (IDC) of P. falciparum. Inhibition of the ubiquitin proteasome system (UPS), a major intracellular proteolytic pathway, effectively reduces infection and parasite replication. P. falciparum and erythrocyte UPS coexist during IDC but the nature of their relationship is largely unknown. We used an approach based on Tandem Ubiquitin-Binding Entities (TUBEs) and 1D gel electrophoresis followed by mass spectrometry to identify major components of the TUBEs-associated ubiquitin proteome of both host and parasite during ring, trophozoite and schizont stages. Ring-exported protein (REX1), a P. falciparum protein located in Maurer's clefts and important for parasite nutrient import, was found to reach a maximum level of ubiquitylation in trophozoites stage. The Homo sapiens (H. sapiens) TUBEs associated ubiquitin proteome decreased during the infection, whereas the equivalent P. falciparum TUBEs-associated ubiquitin proteome counterpart increased. Major cellular processes such as DNA repair, replication, stress response, vesicular transport and catabolic events appear to be regulated by ubiquitylation along the IDC P. falciparum infection. BIOLOGICAL SIGNIFICANCE In this work we analyze for the first time the interconnection between Plasmodium and human red blood cells ubiquitin-regulated proteins in the context of infection. We identified a number of human and Plasmodium proteins whose ubiquitylation pattern changes during the asexual infective stage. We demonstrate that ubiquitylation of REX1, a P. falciparum protein located in Maurer's clefts and important for parasite nutrient import, peaks in trophozoites stage. The ubiquitin-proteome from P. falciparum infected red blood cells (iRBCs) revealed a significant host-parasite crosstalk, underlining the importance of ubiquitin-regulated proteolytic activities during the intraerythrocytic developmental cycle (IDC) of P. falciparum. Major cellular processes defined from gene ontology such as DNA repair, replication, stress response, vesicular transport and catabolic events appear to be regulated by ubiquitylation along the IDC P. falciparum infection. Given the importance of ubiquitylation in the development of infectious diseases, this work provides a number of potential drug-target candidates that should be further explored.
Collapse
Affiliation(s)
- Lydia Mata-Cantero
- Medicines Development Campus, Diseases of the Developing World, GlaxoSmithKline Tres Cantos, Madrid, Spain; Proteomics Platform CICbioGUNE, CIBERehd, ProteoRed-ISCIII, Parque Tecnologico de Bizkaia, Derio, Spain; Ubiquitylation and Cancer Molecular Biology, Inbiomed, San Sebastian, Spain
| | - Mikel Azkargorta
- Proteomics Platform CICbioGUNE, CIBERehd, ProteoRed-ISCIII, Parque Tecnologico de Bizkaia, Derio, Spain
| | - Fabienne Aillet
- Ubiquitylation and Cancer Molecular Biology, Inbiomed, San Sebastian, Spain
| | - Wendy Xolalpa
- Proteomics Platform CICbioGUNE, CIBERehd, ProteoRed-ISCIII, Parque Tecnologico de Bizkaia, Derio, Spain
| | - Maria J LaFuente
- Medicines Development Campus, Diseases of the Developing World, GlaxoSmithKline Tres Cantos, Madrid, Spain
| | - Felix Elortza
- Proteomics Platform CICbioGUNE, CIBERehd, ProteoRed-ISCIII, Parque Tecnologico de Bizkaia, Derio, Spain
| | - Ana Sofia Carvalho
- Computational and Experimental Biology Group, Health Promotion and Chronic Diseases Department, National Institute of Health Dr Ricardo Jorge, Lisbon, Portugal
| | - Julio Martin-Plaza
- Centro de Investigación Básica, GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | - Rune Matthiesen
- Computational and Experimental Biology Group, Health Promotion and Chronic Diseases Department, National Institute of Health Dr Ricardo Jorge, Lisbon, Portugal.
| | - Manuel S Rodriguez
- Proteomics Platform CICbioGUNE, CIBERehd, ProteoRed-ISCIII, Parque Tecnologico de Bizkaia, Derio, Spain; Ubiquitylation and Cancer Molecular Biology, Inbiomed, San Sebastian, Spain; Institut des Technologies Avancées en sciences du Vivant (ITAV), Université de Toulouse, CNRS, UPS, France; University of Toulouse III-Paul Sabatier, 31077 Toulouse, France; Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, France.
| |
Collapse
|
49
|
Structure- and function-based design of Plasmodium-selective proteasome inhibitors. Nature 2016; 530:233-6. [PMID: 26863983 PMCID: PMC4755332 DOI: 10.1038/nature16936] [Citation(s) in RCA: 183] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 12/18/2015] [Indexed: 12/13/2022]
Abstract
The proteasome is a multi-component protease complex responsible for regulating key processes such as the cell cycle and antigen presentation1. Compounds that target the proteasome are potentially valuable tools for the treatment of pathogens that depend on proteasome function for survival and replication. In particular, proteasome inhibitors have been shown to be toxic for the malaria parasite Plasmodium falciparum at all stages of its life cycle2-5. Most compounds that have been tested against the parasite also inhibit the mammalian proteasome resulting in toxicity that precludes their use as therapeutic agents2,6. Therefore, better definition of the substrate specificity and structural properties of the Plasmodium proteasome could enable the development of compounds with sufficient selectivity to allow their use as anti-malarial agents. To accomplish this goal, we used a substrate profiling method to uncover differences in the specificities of the human and P. falciparum proteasome. We designed inhibitors based on amino acid preferences specific to the parasite proteasome, and found that they preferentially inhibit the β 2 subunit. We determined the structure of the P. falciparum 20S proteasome bound to the inhibitor using cryo-electron microscopy (cryo-EM) and single particle analysis, to a resolution of 3.6 Å. These data reveal the unusually open P. falciparum β2 active site and provide valuable information regarding active site architecture that can be used to further refine inhibitor design. Furthermore, consistent with the recent finding that the proteasome is important for stress pathways associated with resistance of artemisinin (ART) family anti-malarials7,8, we observed growth inhibition synergism with low doses of this β 2 selective inhibitor in ART sensitive and resistant parasites. Finally, we demonstrated that a parasite selective inhibitor could be used to attenuate parasite growth in vivo without significant toxicity to the host. Thus, the Plasmodium proteasome is a chemically tractable target that could be exploited by next generation anti-malarial agents.
Collapse
|
50
|
Wang L, Delahunty C, Fritz-Wolf K, Rahlfs S, Helena Prieto J, Yates JR, Becker K. Characterization of the 26S proteasome network in Plasmodium falciparum. Sci Rep 2015; 5:17818. [PMID: 26639022 PMCID: PMC4671066 DOI: 10.1038/srep17818] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 11/06/2015] [Indexed: 11/09/2022] Open
Abstract
In eukaryotic cells, the ubiquitin-proteasome system as a key regulator of protein quality control is an excellent drug target. We therefore aimed to analyze the 26S proteasome complex in the malaria parasite Plasmodium falciparum, which still threatens almost half of the world’s population. First, we established an affinity purification protocol allowing for the isolation of functional 26S proteasome complexes from the parasite. Subunit composition of the proteasome and component stoichiometry were studied and physiologic interacting partners were identified via in situ protein crosslinking. Furthermore, intrinsic ubiquitin receptors of the plasmodial proteasome were determined and their roles in proteasomal substrate recognition were analyzed. Notably, PfUSP14 was characterized as a proteasome-associated deubiquitinase resulting in the concept that targeting proteasomal deubiquitinating activity in P. falciparum may represent a promising antimalarial strategy. The data provide insights into a profound network orchestrated by the plasmodial proteasome and identified novel drug target candidates in the ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Lihui Wang
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, Giessen, Germany
| | - Claire Delahunty
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California
| | | | - Stefan Rahlfs
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, Giessen, Germany
| | - Judith Helena Prieto
- Department of Chemistry, Western Connecticut State University, Danbury, Connecticut
| | - John R Yates
- Max-Planck Institute for Medical Research, Heidelberg, Germany
| | - Katja Becker
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus Liebig University, Giessen, Germany
| |
Collapse
|