1
|
Okombo J, Fidock DA. Towards next-generation treatment options to combat Plasmodium falciparum malaria. Nat Rev Microbiol 2024:10.1038/s41579-024-01099-x. [PMID: 39367132 DOI: 10.1038/s41579-024-01099-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 10/06/2024]
Abstract
Malaria, which is caused by infection of red blood cells with Plasmodium parasites, can be fatal in non-immune individuals if left untreated. The recent approval of the pre-erythrocytic vaccines RTS, S/AS01 and R21/Matrix-M has ushered in hope of substantial reductions in mortality rates, especially when combined with other existing interventions. However, the efficacy of these vaccines is partial, and chemotherapy remains central to malaria treatment and control. For many antimalarial drugs, clinical efficacy has been compromised by the emergence of drug-resistant Plasmodium falciparum strains. Therefore, there is an urgent need for new antimalarial medicines to complement the existing first-line artemisinin-based combination therapies. In this Review, we discuss various opportunities to expand the present malaria treatment space, appraise the current antimalarial drug development pipeline and highlight examples of promising targets. We also discuss other approaches to circumvent antimalarial resistance and how potency against drug-resistant parasites could be retained.
Collapse
Affiliation(s)
- John Okombo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA.
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
2
|
Liu LJ, O'Donoghue AJ, Caffrey CR. The proteasome as a drug target for treatment of parasitic diseases. ADVANCES IN PARASITOLOGY 2024; 126:53-96. [PMID: 39448194 DOI: 10.1016/bs.apar.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
The proteasome is a proteolytically active molecular machine comprising many different protein subunits. It is essential for growth and survival in eukaryotic cells and has long been considered a drug target. Here, we summarize the biology of the proteasome, the early research relating to the development of specific proteasome inhibitors (PIs) for treatment of various cancers, and their translation and eventual evolution as exciting therapies for parasitic diseases. We also highlight the development and adaptation of technologies that have allowed for a deep understanding of the idiosyncrasies of individual parasite proteasomes, as well as the preclinical and clinical advancement of PIs with remarkable therapeutic indices.
Collapse
Affiliation(s)
- Lawrence J Liu
- Center for Discovery and Innovation in Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, United States; Department of Chemistry and Biochemistry, University of California, San Diego, CA, United States.
| | - Anthony J O'Donoghue
- Center for Discovery and Innovation in Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, United States
| | - Conor R Caffrey
- Center for Discovery and Innovation in Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, United States
| |
Collapse
|
3
|
Sojka D, Šnebergerová P. Advances in protease inhibition-based chemotherapy: A decade of insights from Malaria research. ADVANCES IN PARASITOLOGY 2024; 126:205-227. [PMID: 39448191 DOI: 10.1016/bs.apar.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Over the last decade, research on the most studied parasite, Plasmodium falciparum, has disclosed significant findings in protease research. Detailed descriptions of the individual roles of protease isoenzymes from various protease classes encoded by the parasite genome have been elucidated, along with their functional and biochemical characterizations. These insights have enabled the development of innovative chemotherapy using low molecular weight inhibitors targeting specific molecular sites. Progress has been made in understanding the proteolytic cascade associated with the apical complex, particularly the roles of aspartyl proteases plasmepsins IX and X as master regulators. Additionally, advancements in direct and alternative methods of proteasome inhibition and expression regulation have been achieved. Research on digestive/food vacuole-associated proteases, with a focus on essential metalloproteases, has also seen significant developments. The rise of extensive genomic datasets and functional genomic tools for other parasitic organisms now allows these approaches to be applied to the study and treatment of other, less known parasitic diseases, aiming to uncover specific biological mechanisms and develop innovative, less toxic chemotherapies.
Collapse
Affiliation(s)
- Daniel Sojka
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic.
| | - Pavla Šnebergerová
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Ceske Budejovice, Czech Republic; Faculty of Science, University of South Bohemia, Ceske Budejovice, Czech Republic
| |
Collapse
|
4
|
Lawong A, Gahalawat S, Ray S, Ho N, Han Y, Ward KE, Deng X, Chen Z, Kumar A, Xing C, Hosangadi V, Fairhurst KJ, Tashiro K, Liszczak G, Shackleford DM, Katneni K, Chen G, Saunders J, Crighton E, Casas A, Robinson JJ, Imlay LS, Zhang X, Lemoff A, Zhao Z, Angulo-Barturen I, Jiménez-Díaz MB, Wittlin S, Campbell SF, Fidock DA, Laleu B, Charman SA, Ready JM, Phillips MA. Identification of potent and reversible piperidine carboxamides that are species-selective orally active proteasome inhibitors to treat malaria. Cell Chem Biol 2024; 31:1503-1517.e19. [PMID: 39084225 DOI: 10.1016/j.chembiol.2024.07.001] [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: 03/04/2024] [Revised: 05/08/2024] [Accepted: 07/03/2024] [Indexed: 08/02/2024]
Abstract
Malaria remains a global health concern as drug resistance threatens treatment programs. We identified a piperidine carboxamide (SW042) with anti-malarial activity by phenotypic screening. Selection of SW042-resistant Plasmodium falciparum (Pf) parasites revealed point mutations in the Pf_proteasome β5 active-site (Pfβ5). A potent analog (SW584) showed efficacy in a mouse model of human malaria after oral dosing. SW584 had a low propensity to generate resistance (minimum inoculum for resistance [MIR] >109) and was synergistic with dihydroartemisinin. Pf_proteasome purification was facilitated by His8-tag introduction onto β7. Inhibition of Pfβ5 correlated with parasite killing, without inhibiting human proteasome isoforms or showing cytotoxicity. The Pf_proteasome_SW584 cryoelectron microscopy (cryo-EM) structure showed that SW584 bound non-covalently distal from the catalytic threonine, in an unexplored pocket at the β5/β6/β3 subunit interface that has species differences between Pf and human proteasomes. Identification of a reversible, species selective, orally active series with low resistance propensity provides a path for drugging this essential target.
Collapse
Affiliation(s)
- Aloysus Lawong
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Suraksha Gahalawat
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Sneha Ray
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Nhi Ho
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Yan Han
- Department of Biophysics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Kurt E Ward
- Department of Microbiology and Immunology, and Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xiaoyi Deng
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Zhe Chen
- Department of Biophysics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Ashwani Kumar
- Department of Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Chao Xing
- Department of Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA; Department of Bioinformatics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Varun Hosangadi
- Department of Microbiology and Immunology, and Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kate J Fairhurst
- Department of Microbiology and Immunology, and Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Kyuto Tashiro
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Glen Liszczak
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - David M Shackleford
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Kasiram Katneni
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Gong Chen
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Jessica Saunders
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Elly Crighton
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Arturo Casas
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Joshua J Robinson
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Leah S Imlay
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Xiaoyu Zhang
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Andrew Lemoff
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | - Zhiyu Zhao
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Iñigo Angulo-Barturen
- The Art of Discovery, Biscay Science and Technology Park, Astrondo Bidea, BIC Bizkaia Bd 612, Derio, 48160 Bizkaia, Basque Country, Spain
| | - María Belén Jiménez-Díaz
- The Art of Discovery, Biscay Science and Technology Park, Astrondo Bidea, BIC Bizkaia Bd 612, Derio, 48160 Bizkaia, Basque Country, Spain
| | - Sergio Wittlin
- Swiss Tropical and Public Health Institute, 4123 Allschwil, Switzerland; University of Basel Kreuzstrasse 2, 4123 Allschwil, Switzerland
| | | | - David A Fidock
- Department of Microbiology and Immunology, and Columbia University Irving Medical Center, New York, NY 10032, USA; Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Benoît Laleu
- Medicines for Malaria Venture, 1215 Geneva, Switzerland
| | - Susan A Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Joseph M Ready
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA.
| | - Margaret A Phillips
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA.
| |
Collapse
|
5
|
Kabeche S, Meister T, Yeh E. A fast-acting inhibitor of blood-stage P. falciparum with mechanism distinct from artemisinin and chloroquine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607553. [PMID: 39185231 PMCID: PMC11343144 DOI: 10.1101/2024.08.12.607553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Artemisinins are first-line treatment for malaria, prized for their extremely fast reduction of parasite load in patients. New fast-acting antimalarial compounds are urgently needed to counter artemisinin resistance, but the fast parasite reduction observed with artemisinins is rare among antimalarial compounds. Here we show that MMV1580853 has a very fast in vitro killing rate, comparable to that of dihydroartemisinin. Near-complete parasite growth inhibition was observed within 1 hour of treatment with MMV1580853 and dihydroartemisinin, while chloroquine, another fast-acting antimalarial, showed partial growth inhibition after 1h. MMV1580853 was reported to inhibit prenyltransferases, but its fast killing rate is inconsistent with this mechanism-of-action and we were unable to validate any of 3 annotated P. falciparum prenyltransferases as MMV1580853 targets. MMV1580853 also did not phenocopy the inhibition phenotype of either chloroquine or dihydroartemisinin. These results indicate that MMV1580853 has a distinct mechanism-of-action leading to a very fast killing rate. MMV1580853 compound development and investigation of its mechanism-of-action will be critical avenues in the search for drugs matching the remarkable clinical efficacy of artemisinin.
Collapse
Affiliation(s)
- Stephanie Kabeche
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas Meister
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ellen Yeh
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA 94158
| |
Collapse
|
6
|
Withers-Martinez C, Lidumniece E, Hackett F, Collins CR, Taha Z, Blackman MJ, Jirgensons A. Peptidic Boronic Acid Plasmodium falciparum SUB1 Inhibitors with Improved Selectivity over Human Proteasome. J Med Chem 2024; 67:13033-13055. [PMID: 39051854 PMCID: PMC7616463 DOI: 10.1021/acs.jmedchem.4c01005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024]
Abstract
Plasmodium falciparum subtilisin-like serine protease 1 (PfSUB1) is essential for egress of invasive merozoite forms of the parasite, rendering PfSUB1 an attractive antimalarial target. Here, we report studies aimed to improve drug-like properties of peptidic boronic acid PfSUB1 inhibitors including increased lipophilicity and selectivity over human proteasome (H20S). Structure-activity relationship investigations revealed that lipophilic P3 amino acid side chains as well as N-capping groups were well tolerated in retaining PfSUB1 inhibitory potency. At the P1 position, replacing the methyl group with a carboxyethyl substituent led to boralactone PfSUB1 inhibitors with remarkably improved selectivity over H20S. Combining lipophilic end-capping groups with the boralactone reduced the selectivity over H20S. However, compound 4c still showed >60-fold selectivity versus H20S and low nanomolar PfSUB1 inhibitory potency. Importantly, this compound inhibited the growth of a genetically modified P. falciparum line expressing reduced levels of PfSUB1 13-fold more efficiently compared to a wild-type parasite line.
Collapse
Affiliation(s)
| | | | - Fiona Hackett
- Malaria Biochemistry Laboratory, The Francis Crick Institute, LondonNW1 1AT, United Kingdom
| | - Christine R. Collins
- Malaria Biochemistry Laboratory, The Francis Crick Institute, LondonNW1 1AT, United Kingdom
| | - Zahie Taha
- Malaria Biochemistry Laboratory, The Francis Crick Institute, LondonNW1 1AT, United Kingdom
| | - Michael J. Blackman
- Malaria Biochemistry Laboratory, The Francis Crick Institute, LondonNW1 1AT, United Kingdom
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, LondonWC1E 7HT, United Kingdom
| | | |
Collapse
|
7
|
Zhan W, Liu YJ, Kirkman LA, Lin G. Protocol for analysis of intracellular conversion of artezomib molecules into new proteasome inhibitors in Plasmodium falciparum parasites. STAR Protoc 2024; 5:102896. [PMID: 38363687 PMCID: PMC10877166 DOI: 10.1016/j.xpro.2024.102896] [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: 07/20/2023] [Revised: 12/13/2023] [Accepted: 01/31/2024] [Indexed: 02/18/2024] Open
Abstract
Artezomibs (ATZs), dual-pharmacophore molecules comprising of artemisinin and a parasite proteasome inhibitor, hijack parasite ubiquitin proteasome system to transform into new proteasome inhibitors following the activation of artemisinin by heme.1 Here, we present a protocol for using a fluorescent activity-based broad-spectrum proteasome inhibitor probe to study intracellular conversion of ATZ molecules into new proteasome inhibitors in malaria parasites. We describe steps for drug treatment and washout, parasite lysis, proteasome labeling, and visualization. For complete details on the use and execution of this protocol, please refer to Zhan et al.1.
Collapse
Affiliation(s)
- Wenhu Zhan
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Yi Jing Liu
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Laura A Kirkman
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA.
| | - Gang Lin
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
8
|
Pires CV, Cassandra D, Xu S, Laleu B, Burrows JN, Adams JH. Oxidative stress changes the effectiveness of artemisinin in Plasmodium falciparum. mBio 2024; 15:e0316923. [PMID: 38323831 PMCID: PMC10936410 DOI: 10.1128/mbio.03169-23] [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: 12/01/2023] [Accepted: 01/09/2024] [Indexed: 02/08/2024] Open
Abstract
Malaria parasites have adaptive mechanisms to modulate their intracellular redox status to tolerate the enhanced oxidizing effects created by malaria fever, hemoglobinopathies and other stress conditions, including antimalaria drugs. Emerging artemisinin (ART) resistance in Plasmodium falciparum is a complex phenotype linked to the parasite's tolerance of the activated drug's oxidative damage along with changes in vesicular transport, lipid metabolism, DNA repair, and exported proteins. In an earlier study, we discovered that many of these metabolic processes are induced in P. falciparum to respond to the oxidative damage caused by artemisinin, which exhibited a highly significant overlap with the parasite's adaptive response mechanisms to survive febrile temperatures. In addition, there was a significant overlap with the parasite's survival responses to oxidative stress. In this study, we investigated these relationships further using an in vitro model to evaluate if oxidative stress and heat-shock conditions could alter the parasite's response to artemisinin. The results revealed that compared to ideal culture conditions, the antimalarial efficacy of artemisinin was significantly reduced in parasites growing in intraerythrocytic oxidative stress but not in heat-shock condition. In contrast, heat shock significantly reduced the efficacy of lumefantrine that is an important ART combination therapy partner drug. We propose that prolonged exposure to intraerythrocytic microenvironmental oxidative stress, as would occur in endemic regions with high prevalence for sickle trait and other hemoglobinopathies, can predispose malaria parasites to develop tolerance to the oxidative damage caused by antimalarial drugs like artemisinin. IMPORTANCE Emerging resistance to the frontline antimalarial drug artemisinin represents a significant threat to worldwide malaria control and elimination. The patterns of parasite changes associated with emerging resistance represent a complex array of metabolic processes evident in various genetic mutations and altered transcription profiles. Genetic factors identified in regulating P. falciparum sensitivity to artemisinin overlap with the parasite's responses to malarial fever, sickle trait, and other types of oxidative stresses, suggesting conserved inducible survival responses. In this study we show that intraerythrocytic stress conditions, oxidative stress and heat shock, can significantly decrease the sensitivity of the parasite to artemisinin and lumefantrine, respectively. These results indicate that an intraerythrocytic oxidative stress microenvironment and heat-shock condition can alter antimalarial drug efficacy. Evaluating efficacy of antimalarial drugs under ideal in vitro culture conditions may not accurately predict drug efficacy in all malaria patients.
Collapse
Affiliation(s)
- Camilla Valente Pires
- Center for Global Health and Interdisciplinary Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Debora Cassandra
- Center for Global Health and Interdisciplinary Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Shulin Xu
- Center for Global Health and Interdisciplinary Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Benoit Laleu
- Medicines for Malaria Venture, ICC, Geneva, Switzerland
| | | | - John H. Adams
- Center for Global Health and Interdisciplinary Diseases Research and USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
9
|
Jiang Z, Silva EB, Liu C, Fajtová P, Liu LJ, El-Sakkary N, Skinner DE, Syed A, Wang SC, Caffrey CR, O’Donoghue AJ. Development of subunit selective proteasome substrates for Schistosoma species. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.13.580161. [PMID: 38405969 PMCID: PMC10888821 DOI: 10.1101/2024.02.13.580161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Schistosomiasis, or bilharzia, is a neglected tropical disease caused by Schistosoma spp. blood flukes that infects over 200 million people worldwide. Just one partially effective drug is available, and new drugs and drug targets would be welcome. The 20S proteasome is a validated drug target for many parasitic infections, including those caused by Plasmodium and Leishmania. We previously showed that anticancer proteasome inhibitors that act through the Schistosoma mansoni 20S proteasome (Sm20S) kill the parasite in vitro. To advance these initial findings, we employed Multiplex Substrate Profiling by Mass Spectrometry (MSP-MS) to define the substrate cleavage specificities of the three catalytic β subunits of purified Sm20S. The profiles in turn were used to design and synthesize subunit-specific optimized substrates that performed two to eight fold better than the equivalent substrates used to measure the activity of the constitutive human proteasome (c20S). These specific substrates also eliminated the need to purify Sm20S from parasite extracts - a single step enrichment was sufficient to accurately measure substrate hydrolysis and its inhibition with proteasome inhibitors. Finally, we show that the substrate and inhibition profiles for the 20S proteasome from the three medically important schistosome species are similar, suggesting that data arising from an inhibitor development campaign that focuses on Sm20S can be extrapolated to the other two targets with consequent time and cost savings.
Collapse
Affiliation(s)
- Zhenze Jiang
- Center for Discovery and Innovation in Parasitic Disease, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093
| | | | - Chenxi Liu
- Center for Discovery and Innovation in Parasitic Disease, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093
| | - Pavla Fajtová
- Center for Discovery and Innovation in Parasitic Disease, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093
| | - Lawrence J. Liu
- Center for Discovery and Innovation in Parasitic Disease, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093
| | - Nelly El-Sakkary
- Center for Discovery and Innovation in Parasitic Disease, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093
| | - Danielle E. Skinner
- Center for Discovery and Innovation in Parasitic Disease, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093
| | - Ali Syed
- Center for Discovery and Innovation in Parasitic Disease, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093
| | - Steven C Wang
- Center for Discovery and Innovation in Parasitic Disease, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093
| | - Conor R. Caffrey
- Center for Discovery and Innovation in Parasitic Disease, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093
| | - Anthony J. O’Donoghue
- Center for Discovery and Innovation in Parasitic Disease, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093
| |
Collapse
|
10
|
Baygin RC, Yilmaz KC, Acar A. Characterization of dabrafenib-induced drug insensitivity via cellular barcoding and collateral sensitivity to second-line therapeutics. Sci Rep 2024; 14:286. [PMID: 38167959 PMCID: PMC10762103 DOI: 10.1038/s41598-023-50443-3] [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: 05/14/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
Drug insensitivity is arguably one of the biggest challenges in cancer therapeutics. Although effective therapeutic solutions in cancer are limited due to the emergence of drug insensitivity, exploiting evolutionary understanding in this context can provide potential second-line therapeutics sensitizing the drug insensitive populations. Targeted therapeutic agent dabrafenib is used to treat CRC patients with BRAF V600E genotype and insensitivity to dabrafenib is often observed. Understanding underlying clonal architecture of dabrafenib-induced drug insensitivity and identification of potential second-line therapeutics that could sensitize dabrafenib insensitive populations remain to be elucidated. For this purpose, we utilized cellular barcoding technology to decipher dabrafenib-induced clonal evolution in BRAF V600E mutant HT-29 cells. This approach revealed the detection of both pre-existing and de novo barcodes with increased frequencies as a result of dabrafenib insensitivity. Furthermore, our longitudinal monitoring of drug insensitivity based on barcode detection from floating DNA within used medium enabled to identify temporal dynamics of pre-existing and de novo barcodes in relation to dabrafenib insensitivity in HT-29 cells. Moreover, whole-exome sequencing analysis exhibited possible somatic CNVs and SNVs contributing to dabrafenib insensitivity in HT-29 cells. Last, collateral drug sensitivity testing demonstrated oxaliplatin and capecitabine, alone or in combination, as successful second-like therapeutics in inducing collateral sensitivity in dabrafenib-insensitive HT-29 cells. Overall, our findings demonstrate clonal dynamics of dabrafenib-insensitivity in HT-29 cells. In addition, oxaliplatin and capecitabine, alone or in combination, were successful second-line therapeutics in inducing collateral sensitivity in dabrafenib-insensitive HT-29 cells.
Collapse
Affiliation(s)
- Rana Can Baygin
- Department of Biological Sciences, Middle East Technical University, Universiteler Mah. Dumlupınar Bulvarı 1, Çankaya, 06800, Ankara, Turkey
| | - Kubra Celikbas Yilmaz
- Department of Biological Sciences, Middle East Technical University, Universiteler Mah. Dumlupınar Bulvarı 1, Çankaya, 06800, Ankara, Turkey
| | - Ahmet Acar
- Department of Biological Sciences, Middle East Technical University, Universiteler Mah. Dumlupınar Bulvarı 1, Çankaya, 06800, Ankara, Turkey.
| |
Collapse
|
11
|
Hsu HC, Li D, Zhan W, Ye J, Liu YJ, Leung A, Qin J, Crespo B, Gamo FJ, Zhang H, Cui L, Roth A, Kirkman LA, Li H, Lin G. Structures revealing mechanisms of resistance and collateral sensitivity of Plasmodium falciparum to proteasome inhibitors. Nat Commun 2023; 14:8302. [PMID: 38097652 PMCID: PMC10721928 DOI: 10.1038/s41467-023-44077-2] [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: 05/30/2023] [Accepted: 11/29/2023] [Indexed: 12/17/2023] Open
Abstract
The proteasome of the malaria parasite Plasmodium falciparum (Pf20S) is an advantageous drug target because its inhibition kills P. falciparum in multiple stages of its life cycle and synergizes with artemisinins. We recently developed a macrocyclic peptide, TDI-8304, that is highly selective for Pf20S over human proteasomes and is potent in vitro and in vivo against P. falciparum. A mutation in the Pf20S β6 subunit, A117D, confers resistance to TDI-8304, yet enhances both enzyme inhibition and anti-parasite activity of a tripeptide vinyl sulfone β2 inhibitor, WLW-vs. Here we present the high-resolution cryo-EM structures of Pf20S with TDI-8304, of human constitutive proteasome with TDI-8304, and of Pf20Sβ6A117D with WLW-vs that give insights into the species selectivity of TDI-8304, resistance to it, and the collateral sensitivity associated with resistance, including that TDI-8304 binds β2 and β5 in wild type Pf20S as well as WLW-vs binds β2 and β5 in Pf20Sβ6A117D. We further show that TDI-8304 kills P. falciparum as quickly as chloroquine and artemisinin and is active against P. cynomolgi at the liver stage. This increases interest in using these structures to facilitate the development of Pf20S inhibitors that target multiple proteasome subunits and limit the emergence of resistance.
Collapse
Affiliation(s)
- Hao-Chi Hsu
- Department of Structural Biology, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA
| | - Daqiang Li
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Wenhu Zhan
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Jianxiang Ye
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Yi Jing Liu
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Annie Leung
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Junling Qin
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
| | - Benigno Crespo
- Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, 28760, Tres Cantos, Madrid, Spain
| | - Francisco-Javier Gamo
- Global Health Medicines R&D, GlaxoSmithKline, Severo Ochoa 2, 28760, Tres Cantos, Madrid, Spain
| | - Hao Zhang
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Alison Roth
- Department of Drug Discovery, Experimental Therapeutics Branch, The Walter Reed Army Institute of Research, 503 Robert Grant Ave., Silver Spring, 20910, MD, USA
| | - Laura A Kirkman
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Huilin Li
- Department of Structural Biology, Van Andel Institute, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA.
| | - Gang Lin
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
12
|
Imhoff RD, Rosenthal MR, Ashraf K, Bhanot P, Ng CL, Flaherty DP. Identification of covalent fragment inhibitors for Plasmodium falciparum UCHL3 with anti-malarial efficacy. Bioorg Med Chem Lett 2023; 94:129458. [PMID: 37634761 PMCID: PMC10529062 DOI: 10.1016/j.bmcl.2023.129458] [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: 05/26/2023] [Revised: 08/07/2023] [Accepted: 08/22/2023] [Indexed: 08/29/2023]
Abstract
Malaria continues to be a major burden on global health, responsible for 619,000 deaths in 2021. The causative agent of malaria is the eukaryotic parasite Plasmodium. Resistance to artemisinin-based combination therapies (ACTs), the current first-line treatment for malaria, has emerged in Asia, South America, and more recently Africa, where >90% of all malaria-related deaths occur. This has necessitated the identification and investigation of novel parasite proteins and pathways as antimalarial targets, including components of the ubiquitin proteasome system. Here, we investigate Plasmodium falciparum deubiquitinase ubiquitin C-terminal hydrolase L3 (PfUCHL3) as one such target. We carried out a high-throughput screen with covalent fragments and identified seven scaffolds that selectively inhibit the plasmodial UCHL3, but not human UCHL3 or the closely related human UCHL1. After assessing toxicity in human cells, we identified four promising hits and demonstrated their efficacy against asexual P. falciparum blood stages and P. berghei sporozoite stages.
Collapse
Affiliation(s)
- Ryan D Imhoff
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Melissa R Rosenthal
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Kutub Ashraf
- Department of Microbiology, Biochemistry & Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ 07103, United States
| | - Purnima Bhanot
- Department of Microbiology, Biochemistry & Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ 07103, United States
| | - Caroline L Ng
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198, United States; Global Center for Health Security, University of Nebraska Medical Center, Omaha, NE 68198, United States; Department of Biology, University of Omaha, Omaha, NE 68182, United States.
| | - Daniel P Flaherty
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States; Purdue Institute for Drug Discovery, West Lafayette, IN 47907, United States; Purdue Institute of Inflammation, Immunology and Infectious Disease, West Lafayette, IN 47907, United States.
| |
Collapse
|
13
|
Mandt REK, Luth MR, Tye MA, Mazitschek R, Ottilie S, Winzeler EA, Lafuente-Monasterio MJ, Gamo FJ, Wirth DF, Lukens AK. Diverse evolutionary pathways challenge the use of collateral sensitivity as a strategy to suppress resistance. eLife 2023; 12:e85023. [PMID: 37737220 PMCID: PMC10695565 DOI: 10.7554/elife.85023] [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: 11/18/2022] [Accepted: 09/21/2023] [Indexed: 09/23/2023] Open
Abstract
Drug resistance remains a major obstacle to malaria control and eradication efforts, necessitating the development of novel therapeutic strategies to treat this disease. Drug combinations based on collateral sensitivity, wherein resistance to one drug causes increased sensitivity to the partner drug, have been proposed as an evolutionary strategy to suppress the emergence of resistance in pathogen populations. In this study, we explore collateral sensitivity between compounds targeting the Plasmodium dihydroorotate dehydrogenase (DHODH). We profiled the cross-resistance and collateral sensitivity phenotypes of several DHODH mutant lines to a diverse panel of DHODH inhibitors. We focus on one compound, TCMDC-125334, which was active against all mutant lines tested, including the DHODH C276Y line, which arose in selections with the clinical candidate DSM265. In six selections with TCMDC-125334, the most common mechanism of resistance to this compound was copy number variation of the dhodh locus, although we did identify one mutation, DHODH I263S, which conferred resistance to TCMDC-125334 but not DSM265. We found that selection of the DHODH C276Y mutant with TCMDC-125334 yielded additional genetic changes in the dhodh locus. These double mutant parasites exhibited decreased sensitivity to TCMDC-125334 and were highly resistant to DSM265. Finally, we tested whether collateral sensitivity could be exploited to suppress the emergence of resistance in the context of combination treatment by exposing wildtype parasites to both DSM265 and TCMDC-125334 simultaneously. This selected for parasites with a DHODH V532A mutation which were cross-resistant to both compounds and were as fit as the wildtype parent in vitro. The emergence of these cross-resistant, evolutionarily fit parasites highlights the mutational flexibility of the DHODH enzyme.
Collapse
Affiliation(s)
- Rebecca EK Mandt
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public HealthBostonUnited States
| | - Madeline R Luth
- Division of Host Pathogen Systems and Therapeutics, Department of Pediatrics, University of California, San DiegoSan DiegoUnited States
| | - Mark A Tye
- Center for Systems Biology, Massachusetts General HospitalBostonUnited States
- Harvard Graduate School of Arts and SciencesCambridgeUnited States
| | - Ralph Mazitschek
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public HealthBostonUnited States
- Center for Systems Biology, Massachusetts General HospitalBostonUnited States
| | - Sabine Ottilie
- Division of Host Pathogen Systems and Therapeutics, Department of Pediatrics, University of California, San DiegoSan DiegoUnited States
| | - Elizabeth A Winzeler
- Division of Host Pathogen Systems and Therapeutics, Department of Pediatrics, University of California, San DiegoSan DiegoUnited States
- Skaggs School of Pharmaceutical Sciences, University of California, San DiegoLa JollaUnited States
| | | | - Francisco Javier Gamo
- Tres Cantos Medicines Development Campus, Diseases of the Developing World, GlaxoSmithKlineMadridSpain
| | - Dyann F Wirth
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public HealthBostonUnited States
- Infectious Disease and Microbiome Program, The Broad InstituteCambridgeUnited States
| | - Amanda K Lukens
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public HealthBostonUnited States
- Infectious Disease and Microbiome Program, The Broad InstituteCambridgeUnited States
| |
Collapse
|
14
|
Danisik N, Yilmaz KC, Acar A. Identification of collateral sensitivity and evolutionary landscape of chemotherapy-induced drug resistance using cellular barcoding technology. Front Pharmacol 2023; 14:1178489. [PMID: 37497108 PMCID: PMC10366361 DOI: 10.3389/fphar.2023.1178489] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/27/2023] [Indexed: 07/28/2023] Open
Abstract
Background: One of the most significant challenges impeding cancer treatment effectiveness is drug resistance. Combining evolutionary understanding with drug resistance can pave the way for the identification of second-line drug options that can overcome drug resistance. Although capecitabine and irinotecan are commonly used therapeutic agents in the treatment of CRC patients, resistance to these agents is common. The underlying clonal dynamics of resistance to these agents using high-resolution barcode technology and identification of effective second-line drugs in this context remain unclear. Methods and materials: Caco-2 and HT-29 cell lines were barcoded, and then capecitabine and irinotecan resistant derivatives of these cell lines were established. The frequencies of barcodes from resistant cell lines and harvested medium, longitudinally, were determined. Collateral drug sensitivity testing was carried out on resistant Caco-2 and HT-29 cell lines using single agents or drug combinations. The SyngeryFinder tool was used to analyse drug combination testing. Results: In Caco-2 and HT-29 cell lines, barcode frequency measurements revealed clonal dynamics of capecitabine and irinotecan formed by both pre-existing and de novo barcodes, indicating the presence of polyclonal drug resistance. The temporal dynamics of clonal evolution in Caco-2 and HT-29 cell lines were demonstrated by longitudinal analysis of pre-existing and de novo barcodes from harvested medium. In Caco-2 and HT-29 cell lines, collateral drug sensitivity revealed a number of drugs that were effective alone and in combination. Conclusion: The use of barcoding technology reveals the clonal dynamics of chemotherapy-induced drug resistance not only from harvested cell populations, but also from longitudinal sampling throughout the course of clonal evolution. Second-line drugs that sensitize drug-resistant CRC cell lines are identified through collateral drug testing.
Collapse
|
15
|
Florin-Christensen M, Sojka D, Ganzinelli S, Šnebergerová P, Suarez CE, Schnittger L. Degrade to survive: the intricate world of piroplasmid proteases. Trends Parasitol 2023; 39:532-546. [PMID: 37271664 DOI: 10.1016/j.pt.2023.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 06/06/2023]
Abstract
Piroplasmids of the genera Babesia, Theileria, and Cytauxzoon are tick-transmitted parasites with a high impact on animals and humans. They have complex life cycles in their definitive arthropod and intermediate vertebrate hosts involving numerous processes, including invasion of, and egress from, host cells, parasite growth, transformation, and migration. Like other parasitic protozoa, piroplasmids are equipped with different types of protease to fulfill many of such essential processes. Blockade of some key proteases, using inhibitors or antibodies, hinders piroplasmid growth, highlighting their potential usefulness in drug therapies and vaccine development. A better understanding of the functional significance of these enzymes will contribute to the development of improved control measures for the devastating animal and human diseases caused by these pathogens.
Collapse
Affiliation(s)
- Monica Florin-Christensen
- Instituto de Patobiología Veterinaria, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas (CICVyA), INTA-Castelar, Los Reseros y Nicolas Repetto s/n, Hurlingham 1686, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires C1033AAJ, Argentina.
| | - Daniel Sojka
- Institute of Parasitology, Biology Centre, Academy of Sciences of the Czech Republic, Branišovská 1160/31, CZ-37005 České Budějovice, Czech Republic
| | - Sabrina Ganzinelli
- Instituto de Patobiología Veterinaria, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas (CICVyA), INTA-Castelar, Los Reseros y Nicolas Repetto s/n, Hurlingham 1686, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires C1033AAJ, Argentina
| | - Pavla Šnebergerová
- Institute of Parasitology, Biology Centre, Academy of Sciences of the Czech Republic, Branišovská 1160/31, CZ-37005 České Budějovice, Czech Republic; Faculty of Science, University of South Bohemia, CZ-370 05 České Budějovice, Czech Republic
| | - Carlos E Suarez
- Washington State University/Animal Disease Research Unit USDA, Pullman, WA, USA
| | - Leonhard Schnittger
- Instituto de Patobiología Veterinaria, Centro de Investigaciones en Ciencias Veterinarias y Agronómicas (CICVyA), INTA-Castelar, Los Reseros y Nicolas Repetto s/n, Hurlingham 1686, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Ciudad Autónoma de Buenos Aires C1033AAJ, Argentina
| |
Collapse
|
16
|
Pires CV, Oberstaller J, Wang C, Casandra D, Zhang M, Chawla J, Adapa SR, Otto TD, Ferdig MT, Rayner JC, Jiang RHY, Adams JH. Chemogenomic Profiling of a Plasmodium falciparum Transposon Mutant Library Reveals Shared Effects of Dihydroartemisinin and Bortezomib on Lipid Metabolism and Exported Proteins. Microbiol Spectr 2023; 11:e0501422. [PMID: 37067430 PMCID: PMC10269874 DOI: 10.1128/spectrum.05014-22] [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] [Received: 12/06/2022] [Accepted: 03/21/2023] [Indexed: 04/18/2023] Open
Abstract
The antimalarial activity of the frontline drug artemisinin involves generation of reactive oxygen species (ROS) leading to oxidative damage of parasite proteins. To achieve homeostasis and maintain protein quality control in the overwhelmed parasite, the ubiquitin-proteasome system kicks in. Even though molecular markers for artemisinin resistance like pfkelch13 have been identified, the intricate network of mechanisms driving resistance remains to be elucidated. Here, we report a forward genetic screening strategy that enables a broader identification of genetic factors responsible for altering sensitivity to dihydroartemisinin (DHA) and a proteasome inhibitor, bortezomib (BTZ). Using a library of isogenic piggyBac mutants in P. falciparum, we defined phenotype-genotype associations influencing drug responses and highlighted shared mechanisms between the two processes, which mainly included proteasome-mediated degradation and the lipid metabolism genes. Additional transcriptomic analysis of a DHA/BTZ-sensitive piggyBac mutant showed it is possible to find differences between the two response mechanisms on the specific components for regulation of the exportome. Our results provide further insight into the molecular mechanisms of antimalarial drug resistance. IMPORTANCE Malaria control is seriously threatened by the emergence and spread of Plasmodium falciparum resistance to the leading antimalarial, artemisinin. The potent killing activity of artemisinin results from oxidative damage unleashed by free heme activation released by hemoglobin digestion. Although the ubiquitin-proteasome system is considered critical for parasite survival of this toxicity, the diverse genetic changes linked to artemisinin resistance are complex and, so far, have not included the ubiquitin-proteasome system. In this study, we use a systematic forward genetic approach by screening a library of P. falciparum random piggyBac mutants to decipher the genetic factors driving malaria parasite responses to the oxidative stress caused by antimalarial drugs. This study compares phenotype-genotype associations influencing dihydroartemisinin responses with the proteasome inhibitor bortezomib to delineate the role of ubiquitin-proteasome system. Our study highlights shared and unique pathways from the complex array of molecular processes critical for P. falciparum survival resulting from the oxidative damage of artemisinin.
Collapse
Affiliation(s)
- Camilla Valente Pires
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
- USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Jenna Oberstaller
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
- USF Genomics Program, College of Public Health, 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
- USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Debora Casandra
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
- USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Min Zhang
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
- USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Jyotsna Chawla
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
- USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, USA
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, United States
| | - Swamy Rakesh Adapa
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
- USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Thomas D. Otto
- Institute of Infection, Immunity and Inflammation, MVLS, University of Glasgow, Glasgow, United Kingdom
| | - Michael T. Ferdig
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Julian C. Rayner
- Cambridge Institute for Medical Research, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Rays H. Y. Jiang
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
- USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - John H. Adams
- Center for Global Health and Infectious Diseases Research, College of Public Health, University of South Florida, Tampa, Florida, USA
- USF Genomics Program, College of Public Health, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
17
|
Deni I, Stokes BH, Ward KE, Fairhurst KJ, Pasaje CFA, Yeo T, Akbar S, Park H, Muir R, Bick DS, Zhan W, Zhang H, Liu YJ, Ng CL, Kirkman LA, Almaliti J, Gould AE, Duffey M, O'Donoghue AJ, Uhlemann AC, Niles JC, da Fonseca PCA, Gerwick WH, Lin G, Bogyo M, Fidock DA. Mitigating the risk of antimalarial resistance via covalent dual-subunit inhibition of the Plasmodium proteasome. Cell Chem Biol 2023; 30:470-485.e6. [PMID: 36963402 PMCID: PMC10198959 DOI: 10.1016/j.chembiol.2023.03.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/10/2023] [Accepted: 03/02/2023] [Indexed: 03/26/2023]
Abstract
The Plasmodium falciparum proteasome constitutes a promising antimalarial target, with multiple chemotypes potently and selectively inhibiting parasite proliferation and synergizing with the first-line artemisinin drugs, including against artemisinin-resistant parasites. We compared resistance profiles of vinyl sulfone, epoxyketone, macrocyclic peptide, and asparagine ethylenediamine inhibitors and report that the vinyl sulfones were potent even against mutant parasites resistant to other proteasome inhibitors and did not readily select for resistance, particularly WLL that displays covalent and irreversible binding to the catalytic β2 and β5 proteasome subunits. We also observed instances of collateral hypersensitivity, whereby resistance to one inhibitor could sensitize parasites to distinct chemotypes. Proteasome selectivity was confirmed using CRISPR/Cas9-edited mutant and conditional knockdown parasites. Molecular modeling of proteasome mutations suggested spatial contraction of the β5 P1 binding pocket, compromising compound binding. Dual targeting of P. falciparum proteasome subunits using covalent inhibitors provides a potential strategy for restoring artemisinin activity and combating the spread of drug-resistant malaria.
Collapse
Affiliation(s)
- Ioanna Deni
- Center for Malaria Therapeutics and Antimicrobial Resistance and Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Barbara H Stokes
- Center for Malaria Therapeutics and Antimicrobial Resistance and Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Kurt E Ward
- Center for Malaria Therapeutics and Antimicrobial Resistance and Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Kate J Fairhurst
- Center for Malaria Therapeutics and Antimicrobial Resistance and Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Tomas Yeo
- Center for Malaria Therapeutics and Antimicrobial Resistance and Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Shirin Akbar
- School of Molecular Biosciences, University of Glasgow, Glasgow, Scotland, UK
| | - Heekuk Park
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Ryan Muir
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniella S Bick
- Center for Malaria Therapeutics and Antimicrobial Resistance and Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Wenhu Zhan
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Hao Zhang
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Yi Jing Liu
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Caroline L Ng
- Global Center for Health Security, University of Nebraska Medical Center, Omaha, NE, USA; Department of Biology, University of Nebraska Omaha, Omaha, NE, USA; Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Laura A Kirkman
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Jehad Almaliti
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, USA; Department of Pharmaceutical Sciences, College of Pharmacy, The University of Jordan, Amman, Jordan
| | | | | | - Anthony J O'Donoghue
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Anne-Catrin Uhlemann
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Jacquin C Niles
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - William H Gerwick
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, USA; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Gang Lin
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - David A Fidock
- Center for Malaria Therapeutics and Antimicrobial Resistance and Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
18
|
Xiao Z, Gray JL, Tate EW. Weaponizing the proteasome to overcome antimalarial drug resistance. Cell Chem Biol 2023; 30:415-417. [PMID: 37207629 DOI: 10.1016/j.chembiol.2023.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 04/24/2023] [Accepted: 04/24/2023] [Indexed: 05/21/2023]
Abstract
In this issue of Cell Chemical Biology, Zhan et al. report dual-pharmacophore molecules ("artezomibs"), combining an artemisinin and proteasome inhibitor that exhibit potent activity against both wild-type and drug-resistant malarial parasites.1 This study indicates that artezomibs offer a promising approach to combat drug resistance encountered by current antimalarial therapies.
Collapse
Affiliation(s)
- Zhangping Xiao
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London W12 0BZ, UK
| | - Janine L Gray
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London W12 0BZ, UK
| | - Edward W Tate
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City Campus, Wood Lane, London W12 0BZ, UK.
| |
Collapse
|
19
|
Zhan W, Li D, Subramanyaswamy SB, Liu YJ, Yang C, Zhang H, Harris JC, Wang R, Zhu S, Rocha H, Sherman J, Qin J, Herring M, Simwela NV, Waters AP, Sukenick G, Cui L, Rodriguez A, Deng H, Nathan CF, Kirkman LA, Lin G. Dual-pharmacophore artezomibs hijack the Plasmodium ubiquitin-proteasome system to kill malaria parasites while overcoming drug resistance. Cell Chem Biol 2023; 30:457-469.e11. [PMID: 37148884 PMCID: PMC10240386 DOI: 10.1016/j.chembiol.2023.04.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/02/2023] [Accepted: 04/06/2023] [Indexed: 05/08/2023]
Abstract
Artemisinins (ART) are critical anti-malarials and despite their use in combination therapy, ART-resistant Plasmodium falciparum is spreading globally. To counter ART resistance, we designed artezomibs (ATZs), molecules that link an ART with a proteasome inhibitor (PI) via a non-labile amide bond and hijack parasite's own ubiquitin-proteasome system to create novel anti-malarials in situ. Upon activation of the ART moiety, ATZs covalently attach to and damage multiple parasite proteins, marking them for proteasomal degradation. When damaged proteins enter the proteasome, their attached PIs inhibit protease function, potentiating the parasiticidal action of ART and overcoming ART resistance. Binding of the PI moiety to the proteasome active site is enhanced by distal interactions of the extended attached peptides, providing a mechanism to overcome PI resistance. ATZs have an extra mode of action beyond that of each component, thereby overcoming resistance to both components, while avoiding transient monotherapy seen when individual agents have disparate pharmacokinetic profiles.
Collapse
Affiliation(s)
- Wenhu Zhan
- Department of Microbiology and Immunology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Daqiang Li
- Department of Microbiology and Immunology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | | | - Yi Jing Liu
- Department of Medicine, Division of Infectious Diseases, 1300 York Avenue, New York, NY 10065, USA
| | - Changmei Yang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Hao Zhang
- Department of Microbiology and Immunology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Jacob C Harris
- Department of Medicine, Division of Infectious Diseases, 1300 York Avenue, New York, NY 10065, USA
| | - Rong Wang
- NMR Analytical Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Songbiao Zhu
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Hedy Rocha
- Division of Parasitology, Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | - Julian Sherman
- Division of Parasitology, Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | - Junling Qin
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Mikayla Herring
- Department of Medicine, Division of Infectious Diseases, 1300 York Avenue, New York, NY 10065, USA
| | - Nelson V Simwela
- School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - Andrew P Waters
- School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - George Sukenick
- NMR Analytical Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Liwang Cui
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Ana Rodriguez
- Division of Parasitology, Department of Microbiology, New York University School of Medicine, New York, NY, USA
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systematic Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Carl F Nathan
- Department of Microbiology and Immunology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Laura A Kirkman
- Department of Microbiology and Immunology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Department of Medicine, Division of Infectious Diseases, 1300 York Avenue, New York, NY 10065, USA.
| | - Gang Lin
- Department of Microbiology and Immunology, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
20
|
Ware KE, Thomas BC, Olawuni PD, Sheth MU, Hawkey N, Yeshwanth M, Miller BC, Vietor KJ, Jolly MK, Kim SY, Armstrong AJ, Somarelli JA. A synthetic lethal screen for Snail-induced enzalutamide resistance identifies JAK/STAT signaling as a therapeutic vulnerability in prostate cancer. Front Mol Biosci 2023; 10:1104505. [PMID: 37228586 PMCID: PMC10203420 DOI: 10.3389/fmolb.2023.1104505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Despite substantial improvements in the treatment landscape of prostate cancer, the evolution of hormone therapy-resistant and metastatic prostate cancer remains a major cause of cancer-related death globally. The mainstay of treatment for advanced prostate cancer is targeting of androgen receptor signaling, including androgen deprivation therapy plus second-generation androgen receptor blockade (e.g., enzalutamide, apalutamide, darolutamide), and/or androgen synthesis inhibition (abiraterone). While these agents have significantly prolonged the lives of patients with advanced prostate cancer, is nearly universal. This therapy resistance is mediated by diverse mechanisms, including both androgen receptor-dependent mechanisms, such as androgen receptor mutations, amplifications, alternative splicing, and amplification, as well as non-androgen receptor-mediated mechanisms, such as lineage plasticity toward neuroendocrine-like or epithelial-mesenchymal transition (EMT)-like lineages. Our prior work identified the EMT transcriptional regulator Snail as critical to hormonal therapy resistance and is commonly detected in human metastatic prostate cancer. In the current study, we sought to interrogate the actionable landscape of EMT-mediated hormone therapy resistant prostate cancer to identify synthetic lethality and collateral sensitivity approaches to treating this aggressive, therapy-resistant disease state. Using a combination of high-throughput drug screens and multi-parameter phenotyping by confluence imaging, ATP production, and phenotypic plasticity reporters of EMT, we identified candidate synthetic lethalities to Snail-mediated EMT in prostate cancer. These analyses identified multiple actionable targets, such as XPO1, PI3K/mTOR, aurora kinases, c-MET, polo-like kinases, and JAK/STAT as synthetic lethalities in Snail+ prostate cancer. We validated these targets in a subsequent validation screen in an LNCaP-derived model of resistance to sequential androgen deprivation and enzalutamide. This follow-up screen provided validation of inhibitors of JAK/STAT and PI3K/mTOR as therapeutic vulnerabilities for both Snail+ and enzalutamide-resistant prostate cancer.
Collapse
Affiliation(s)
- Kathryn E. Ware
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, United States
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University Medical Center, Durham, NC, United States
| | - Beatrice C. Thomas
- Dr. Kiran C Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Pelumi D. Olawuni
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, United States
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University Medical Center, Durham, NC, United States
| | - Maya U. Sheth
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, United States
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University Medical Center, Durham, NC, United States
| | - Nathan Hawkey
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, United States
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University Medical Center, Durham, NC, United States
| | - M. Yeshwanth
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Brian C. Miller
- Division of Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Katherine J. Vietor
- Division of Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - So Young Kim
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, United States
| | - Andrew J. Armstrong
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, United States
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University Medical Center, Durham, NC, United States
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, United States
| | - Jason A. Somarelli
- Department of Medicine, Division of Medical Oncology, Duke University Medical Center, Durham, NC, United States
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Duke University Medical Center, Durham, NC, United States
| |
Collapse
|
21
|
Almaliti J, Fajtová P, Calla J, LaMonte GM, Feng M, Rocamora F, Ottilie S, Glukhov E, Boura E, Suhandynata RT, Momper JD, Gilson MK, Winzeler EA, Gerwick WH, O'Donoghue AJ. Development of Potent and Highly Selective Epoxyketone-Based Plasmodium Proteasome Inhibitors. Chemistry 2023; 29:e202203958. [PMID: 36617500 PMCID: PMC10894724 DOI: 10.1002/chem.202203958] [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: 12/19/2022] [Revised: 01/03/2023] [Accepted: 01/03/2023] [Indexed: 01/10/2023]
Abstract
Here, we present remarkable epoxyketone-based proteasome inhibitors with low nanomolar in vitro potency for blood-stage Plasmodium falciparum and low cytotoxicity for human cells. Our best compound has more than 2,000-fold greater selectivity for erythrocytic-stage P. falciparum over HepG2 and H460 cells, which is largely driven by the accommodation of the parasite proteasome for a D-amino acid in the P3 position and the preference for a difluorobenzyl group in the P1 position. We isolated the proteasome from P. falciparum cell extracts and determined that the best compound is 171-fold more potent at inhibiting the β5 subunit of P. falciparum proteasome when compared to the same subunit of the human constitutive proteasome. These compounds also significantly reduce parasitemia in a P. berghei mouse infection model and prolong survival of animals by an average of 6 days. The current epoxyketone inhibitors are ideal starting compounds for orally bioavailable anti-malarial drugs.
Collapse
Affiliation(s)
- Jehad Almaliti
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
- Department Pharmaceutical Sciences, College of Pharmacy, University of Jordan, Amman, 11942, Jordan
| | - Pavla Fajtová
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 16610, Prague, Czech Republic
| | - Jaeson Calla
- Department of Pediatrics, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Gregory M LaMonte
- Department of Pediatrics, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Mudong Feng
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Frances Rocamora
- Department of Pediatrics, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Sabine Ottilie
- Department of Pediatrics, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
- Calibr, a division of The Scripps Research Institute, 11119 N Torrey Pines Rd, La Jolla, California, 92093, USA
| | - Evgenia Glukhov
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Evzen Boura
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 16610, Prague, Czech Republic
| | - Raymond T Suhandynata
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Jeremiah D Momper
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Michael K Gilson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Elizabeth A Winzeler
- Department of Pediatrics, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - William H Gerwick
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Anthony J O'Donoghue
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| |
Collapse
|
22
|
Bopp S, Pasaje CFA, Summers RL, Magistrado-Coxen P, Schindler KA, Corpas-Lopez V, Yeo T, Mok S, Dey S, Smick S, Nasamu AS, Demas AR, Milne R, Wiedemar N, Corey V, Gomez-Lorenzo MDG, Franco V, Early AM, Lukens AK, Milner D, Furtado J, Gamo FJ, Winzeler EA, Volkman SK, Duffey M, Laleu B, Fidock DA, Wyllie S, Niles JC, Wirth DF. Potent acyl-CoA synthetase 10 inhibitors kill Plasmodium falciparum by disrupting triglyceride formation. Nat Commun 2023; 14:1455. [PMID: 36927839 PMCID: PMC10020447 DOI: 10.1038/s41467-023-36921-2] [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: 06/05/2020] [Accepted: 02/20/2023] [Indexed: 03/18/2023] Open
Abstract
Identifying how small molecules act to kill malaria parasites can lead to new "chemically validated" targets. By pressuring Plasmodium falciparum asexual blood stage parasites with three novel structurally-unrelated antimalarial compounds (MMV665924, MMV019719 and MMV897615), and performing whole-genome sequence analysis on resistant parasite lines, we identify multiple mutations in the P. falciparum acyl-CoA synthetase (ACS) genes PfACS10 (PF3D7_0525100, M300I, A268D/V, F427L) and PfACS11 (PF3D7_1238800, F387V, D648Y, and E668K). Allelic replacement and thermal proteome profiling validates PfACS10 as a target of these compounds. We demonstrate that this protein is essential for parasite growth by conditional knockdown and observe increased compound susceptibility upon reduced expression. Inhibition of PfACS10 leads to a reduction in triacylglycerols and a buildup of its lipid precursors, providing key insights into its function. Analysis of the PfACS11 gene and its mutations point to a role in mediating resistance via decreased protein stability.
Collapse
Affiliation(s)
- Selina Bopp
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, The Broad Institute, Cambridge, MA, USA
| | | | - Robert L Summers
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, The Broad Institute, Cambridge, MA, USA
| | - Pamela Magistrado-Coxen
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, The Broad Institute, Cambridge, MA, USA
| | - Kyra A Schindler
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Victoriano Corpas-Lopez
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Tomas Yeo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Sachel Mok
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Sumanta Dey
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sebastian Smick
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Armiyaw S Nasamu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Allison R Demas
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, The Broad Institute, Cambridge, MA, USA
| | - Rachel Milne
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Natalie Wiedemar
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Victoria Corey
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA, USA
| | - Maria De Gracia Gomez-Lorenzo
- Tres Cantos Medicines Research and Development Campus, Diseases of the Developing World, GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | - Virginia Franco
- Tres Cantos Medicines Research and Development Campus, Diseases of the Developing World, GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | - Angela M Early
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, The Broad Institute, Cambridge, MA, USA
| | - Amanda K Lukens
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, The Broad Institute, Cambridge, MA, USA
| | - Danny Milner
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, The Broad Institute, Cambridge, MA, USA
| | - Jeremy Furtado
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Francisco-Javier Gamo
- Tres Cantos Medicines Research and Development Campus, Diseases of the Developing World, GlaxoSmithKline, Tres Cantos, Madrid, Spain
| | - Elizabeth A Winzeler
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Sarah K Volkman
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, The Broad Institute, Cambridge, MA, USA
- College of Natural, Behavioral, and Health Sciences, Simmons University, Boston, MA, USA
| | | | - Benoît Laleu
- Medicines for Malaria Venture, Geneva, Switzerland
| | - David A Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Susan Wyllie
- Wellcome Centre for Anti-Infectives Research, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Jacquin C Niles
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Dyann F Wirth
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Infectious Disease and Microbiome Program, The Broad Institute, Cambridge, MA, USA.
| |
Collapse
|
23
|
A Plasmodium falciparum RING Finger E3 Ubiquitin Ligase Modifies the Roles of PfMDR1 and PfCRT in Parasite Drug Responses. Antimicrob Agents Chemother 2023; 67:e0082122. [PMID: 36625569 PMCID: PMC9933707 DOI: 10.1128/aac.00821-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Protein ubiquitination is an important posttranslational regulation mechanism that mediates Plasmodium development and modifies parasite responses to antimalarial drugs. Although mutations in several parasite ubiquitination enzymes have been linked to increased drug tolerance, the molecular mechanisms by which ubiquitination pathways mediate these parasite responses remain largely unknown. Here, we investigate the roles of a Plasmodium falciparum ring finger ubiquitin ligase (PfRFUL) in parasite development and in responses to antimalarial drugs. We engineered a transgenic parasite having the Pfrful gene tagged with an HA-2A-NeoR-glmS sequence to knockdown (KD) Pfrful expression using glucosamine (GlcN). A Western blot analysis of the proteins from GlcN-treated pSLI-HA-NeoR-glmS-tagged (PfRFULg) parasites, relative to their wild-type (Dd2) controls, showed changes in the ubiquitination of numerous proteins. PfRFUL KD rendered the parasites more sensitive to multiple antimalarial drugs, including mefloquine, piperaquine, amodiaquine, and dihydroartemisinin. PfRFUL KD also decreased the protein level of the P. falciparum multiple drug resistance 1 protein (PfMDR1) and altered the ratio of two bands of the P. falciparum chloroquine resistance transporter (PfCRT), suggesting contributions to the changed drug responses by the altered ubiquitination of these two molecules. The inhibition of proteasomal protein degradation by epoxomicin increased the PfRFUL level, suggesting the degradation of PfRFUL by the proteasome pathways, whereas the inhibition of E3 ubiquitin ligase activities by JNJ26854165 reduced the PfRFUL level. This study reveals the potential mechanisms of PfRFUL in modifying the expression of drug transporters and their roles in parasite drug responses. PfRFUL could be a potential target for antimalarial drug development.
Collapse
|
24
|
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
|
25
|
Zhan W, Li D, Saha P, Wang R, Zhang H, Ajay AK, Deban C, Sukenick G, Azzi J, Lin G. Discovery of Highly Selective Inhibitors of the Human Constitutive Proteasome β5c Chymotryptic Subunit. J Med Chem 2023; 66:1172-1185. [PMID: 36608337 PMCID: PMC10157300 DOI: 10.1021/acs.jmedchem.2c00733] [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] [Indexed: 01/07/2023]
Abstract
We describe our discovery and development of potent and highly selective inhibitors of human constitutive proteasome chymotryptic activity (β5c). Structure-activity relationship studies of the novel class of inhibitors focused on optimization of N-cap, C-cap, and side chain of the chemophore asparagine. Compound 32 is the most potent and selective β5c inhibitor in this study. A docking study provides a structure rationale for potency and selectivity. Kinetic studies show a reversible and noncompetitive inhibition mechanism. It enters the cells to engage the proteasome target, potently and selectively kills multiple myeloma cells, and does so by synergizing with a β5i-selective inhibitor.
Collapse
Affiliation(s)
- Wenhu Zhan
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, United States
| | - Daqiang Li
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, United States
| | - Priya Saha
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, United States
| | - Rong Wang
- NMR Analytical Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Hao Zhang
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, United States
| | - Amrendra K. Ajay
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Christa Deban
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - George Sukenick
- NMR Analytical Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Jamil Azzi
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Gang Lin
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, NY 10065, United States
| |
Collapse
|
26
|
Lai JW, Maah MJ, Tan KW, Sarip R, Lim YAL, Ganguly R, Khaw LT, Ng CH. Dinuclear and mononuclear metal(II) polypyridyl complexes against drug-sensitive and drug-resistant Plasmodium falciparum and their mode of action. Malar J 2022; 21:386. [PMID: 36528584 PMCID: PMC9758846 DOI: 10.1186/s12936-022-04406-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Malaria remains one of the most virulent and deadliest parasitic disease in the world, particularly in Africa and Southeast Asia. Widespread occurrence of artemisinin-resistant Plasmodium falciparum strains from the Greater Mekong Subregion is alarming. This hinders the national economies, as well as being a major drawback in the effective control and elimination of malaria worldwide. Clearly, an effective anti-malarial drug is urgently needed. METHODS The dinuclear and mononuclear copper(II) and zinc(II) complexes were synthesized in ethanolic solution and characterized by various physical measurements (FTIR, CHN elemental analysis, solubility, ESI-MS, UV-Visible, conductivity and magnetic moment, and NMR). X-ray crystal structure of the dicopper(II) complex was determined. The in vitro haemolytic activities of these metal complexes were evaluated spectroscopically on B+ blood while the anti-malarial potency was performed in vitro on blood stage drug-sensitive Plasmodium falciparum 3D7 (Pf3D7) and artemisinin-resistant Plasmodium falciparum IPC5202 (Pf5202) with fluorescence dye. Mode of action of metal complexes were conducted to determine the formation of reactive oxygen species using PNDA and DCFH-DA dyes, JC-1 depolarization of mitochondrial membrane potential, malarial 20S proteasome inhibition with parasite lysate, and morphological studies using Giemsa and Hoechst stains. RESULTS Copper(II) complexes showed anti-malarial potency against both Pf3D7 and Pf5202 in sub-micromolar to micromolar range. The zinc(II) complexes were effective against Pf3D7 with excellent therapeutic index but encountered total resistance against Pf5202. Among the four, the dinuclear copper(II) complex was the most potent against both strains. The zinc(II) complexes caused no haemolysis of RBC while copper(II) complexes induced increased haemolysis with increasing concentration. Further mechanistic studies of both copper(II) complexes on both Pf3D7 and Pf5202 strains showed induction of ROS, 20S malarial proteasome inhibition, loss of mitochondrial membrane potential and morphological features indicative of apoptosis. CONCLUSION The dinuclear [Cu(phen)-4,4'-bipy-Cu(phen)](NO3)4 is highly potent and can overcome the total drug-resistance of Pf5202 towards chloroquine and artemisinin. The other three copper(II) and zinc(II) complexes were only effective towards the drug-sensitive Pf3D7, with the latter causing no haemolysis of RBC. Their mode of action involves multiple targets.
Collapse
Affiliation(s)
- Jing Wei Lai
- grid.10347.310000 0001 2308 5949Department of Chemistry, Faculty of Science, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Mohd Jamil Maah
- grid.10347.310000 0001 2308 5949Department of Chemistry, Faculty of Science, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Kong Wai Tan
- grid.10347.310000 0001 2308 5949Department of Chemistry, Faculty of Science, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Rozie Sarip
- grid.10347.310000 0001 2308 5949Department of Chemistry, Faculty of Science, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Yvonne Ai Lian Lim
- grid.10347.310000 0001 2308 5949Department of Parasitology, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Rakesh Ganguly
- grid.410868.30000 0004 1781 342XShiv Nadar University, Greater Noida, India
| | - Loke Tim Khaw
- grid.411729.80000 0000 8946 5787Department of Microbiology and Immunology, School of Medicine, International Medical University, 57000 Kuala Lumpur, Malaysia
| | - Chew Hee Ng
- grid.411729.80000 0000 8946 5787Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, 57000 Kuala Lumpur, Malaysia
| |
Collapse
|
27
|
Safe, Effective, and Inexpensive Clearance of Mycoplasma Contamination from Cultures of Apicomplexan Parasites with Sparfloxacin. Microbiol Spectr 2022; 10:e0349722. [PMID: 36190416 PMCID: PMC9603166 DOI: 10.1128/spectrum.03497-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Most commercial products cannot be used for clearance of Mycoplasma contamination from cultures of apicomplexan parasites due to the parasites' dependence on the apicoplast, an essential organelle with DNA replication and translation machinery of cyanobacterial origin. The lone exception, mycoplasma removal agent (MRA), is relatively expensive, and some mycoplasma strains have shown resistance to clearance with MRA. Here, we report that the fluoroquinolone antibiotic sparfloxacin is a safe, effective, and inexpensive alternative for treatment of mycoplasma contamination in cultures of apicomplexan parasites. Sparfloxacin cleared both MRA-sensitive and MRA-resistant mycoplasma species from P. falciparum cultures at 1 and 4 μg/mL, respectively. We show that cultures of three different apicomplexan parasites can be maintained at concentrations of sparfloxacin required to clear mycoplasma without resulting in substantial deleterious effects on parasite growth. We also describe an alternative low-cost, in-house PCR assay for detecting mycoplasma. These findings will be useful to laboratories maintaining apicomplexan parasites in vitro, especially in low-resource environments, where the high cost of commercial products creates an economic barrier for detecting and eliminating mycoplasma from culture. IMPORTANCE These findings will be useful to laboratories maintaining apicomplexan parasites in vitro, especially in low-resource environments, where the high cost of commercial products creates an economic barrier for detecting and eliminating Mycoplasma from culture.
Collapse
|
28
|
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
|
29
|
Zhang H, Ginn J, Zhan W, Liu YJ, Leung A, Toita A, Okamoto R, Wong TT, Imaeda T, Hara R, Yukawa T, Michino M, Vendome J, Beuming T, Sato K, Aso K, Meinke PT, Nathan CF, Kirkman LA, Lin G. Design, Synthesis, and Optimization of Macrocyclic Peptides as Species-Selective Antimalaria Proteasome Inhibitors. J Med Chem 2022; 65:9350-9375. [PMID: 35727231 PMCID: PMC10152543 DOI: 10.1021/acs.jmedchem.2c00611] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
With over 200 million cases and close to half a million deaths each year, malaria is a threat to global health, particularly in developing countries. Plasmodium falciparum, the parasite that causes the most severe form of the disease, has developed resistance to all antimalarial drugs. Resistance to the first-line antimalarial artemisinin and to artemisinin combination therapies is widespread in Southeast Asia and is emerging in sub-Saharan Africa. The P. falciparum proteasome is an attractive antimalarial target because its inhibition kills the parasite at multiple stages of its life cycle and restores artemisinin sensitivity in parasites that have become resistant through mutation in Kelch K13. Here, we detail our efforts to develop noncovalent, macrocyclic peptide malaria proteasome inhibitors, guided by structural analysis and pharmacokinetic properties, leading to a potent, species-selective, metabolically stable inhibitor.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, New York 10065, United States
| | - John Ginn
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Wenhu Zhan
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, New York 10065, United States
| | - Yi J Liu
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, New York 10065, United States
| | - Annie Leung
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, 1300 York Ave., New York, New York 10065, United States
| | - Akinori Toita
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Rei Okamoto
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Tzu-Tshin Wong
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Toshihiro Imaeda
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Ryoma Hara
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Takafumi Yukawa
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Mayako Michino
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | | | - Thijs Beuming
- Schrödinger, Inc., New York, New York 10036, United States
| | - Kenjiro Sato
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Kazuyoshi Aso
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Peter T Meinke
- Tri-Institutional Therapeutics Discovery Institute, 413 E. 69th St, New York, New York 10065, United States
| | - Carl F Nathan
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, New York 10065, United States
| | - Laura A Kirkman
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, New York 10065, United States.,Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, 1300 York Ave., New York, New York 10065, United States
| | - Gang Lin
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave., New York, New York 10065, United States
| |
Collapse
|
30
|
Ignatz-Hoover JJ, Murphy EV, Driscoll JJ. Targeting Proteasomes in Cancer and Infectious Disease: A Parallel Strategy to Treat Malignancies and Microbes. Front Cell Infect Microbiol 2022; 12:925804. [PMID: 35873166 PMCID: PMC9302482 DOI: 10.3389/fcimb.2022.925804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/02/2022] [Indexed: 02/05/2023] Open
Abstract
Essential core pathways of cellular biology are preserved throughout evolution, highlighting the importance of these pathways for both bacteria and human cancer cells alike. Cell viability requires a proper balance between protein synthesis and degradation in order to maintain integrity of the proteome. Proteasomes are highly intricate, tightly regulated multisubunit complexes that are critical to achieve protein homeostasis (proteostasis) through the selective degradation of misfolded, redundant and damaged proteins. Proteasomes function as the catalytic core of the ubiquitin-proteasome pathway (UPP) which regulates a myriad of essential processes including growth, survival, differentiation, drug resistance and apoptosis. Proteasomes recognize and degrade proteins that have been marked by covalently attached poly-ubiquitin chains. Deregulation of the UPP has emerged as an essential etiology of many prominent diseases, including cancer. Proteasome inhibitors selectively target cancer cells, including those resistant to chemotherapy, while sparing healthy cells. Proteasome inhibition has emerged as a transformative anti-myeloma strategy that has extended survival for certain patient populations from 3 to 8 years. The structural architecture and functional activity of proteasomes is conserved from Archaea to humans to support the concept that proteasomes are actionable targets that can be inhibited in pathogenic organisms to improve the treatment of infectious diseases. Proteasomes have an essential role during all stages of the parasite life cycle and features that distinguish proteasomes in pathogens from human forms have been revealed. Advancement of inhibitors that target Plasmodium and Mycobacterial proteasomes is a means to improve treatment of malaria and tuberculosis. In addition, PIs may also synergize with current frontline agents support as resistance to conventional drugs continues to increase. The proteasome represents a highly promising, actionable target to combat infectious diseases that devastate lives and livelihoods around the globe.
Collapse
Affiliation(s)
- James J. Ignatz-Hoover
- Division of Hematology & Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Adult Hematologic Malignancies & Stem Cell Transplant Section, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Elena V. Murphy
- Case Western Reserve University, Department of Biochemistry, Cleveland, OH, United States
| | - James J. Driscoll
- Division of Hematology & Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Adult Hematologic Malignancies & Stem Cell Transplant Section, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
31
|
Stochastic expression of invasion genes in Plasmodium falciparum schizonts. Nat Commun 2022; 13:3004. [PMID: 35637187 PMCID: PMC9151791 DOI: 10.1038/s41467-022-30605-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/22/2022] [Indexed: 12/15/2022] Open
Abstract
Genetically identical cells are known to exhibit differential phenotypes in the same environmental conditions. These phenotypic variants are linked to transcriptional stochasticity and have been shown to contribute towards adaptive flexibility of a wide range of unicellular organisms. Here, we investigate transcriptional heterogeneity and stochastic gene expression in Plasmodium falciparum by performing the quasilinear multiple annealing and looping based amplification cycles (MALBAC) based amplification and single cell RNA sequencing of blood stage schizonts. Our data reveals significant transcriptional variations in the schizont stage with a distinct group of highly variable invasion gene transcripts being identified. Moreover, the data reflects several diversification processes including putative developmental “checkpoint”; transcriptomically distinct parasite sub-populations and transcriptional switches in variable gene families (var, rifin, phist). Most of these features of transcriptional variability are preserved in isogenic parasite cell populations (albeit with a lesser amplitude) suggesting a role of epigenetic factors in cell-to-cell transcriptional variations in human malaria parasites. Lastly, we apply quantitative RT-PCR and RNA-FISH approach and confirm stochastic expression of key invasion genes, such as, msp1, msp3, msp7, eba181 and ama1 which represent prime candidates for invasion-blocking vaccines. Genetically identical cells can be phenotypically diverse to allow adaptive flexibility in a given environment. This phenotypic diversity is driven by epigenetic and transcriptional variability. Here, Tripathi et al. perform scRNA-seq of isogenic and non-isogenic Plasmodium falciparum schizont populations to explore transcriptional heterogeneity and stochastic gene expression during the course of development.
Collapse
|
32
|
Small-Saunders JL, Hagenah LM, Wicht KJ, Dhingra SK, Deni I, Kim J, Vendome J, Gil-Iturbe E, Roepe PD, Mehta M, Mancia F, Quick M, Eppstein MJ, Fidock DA. Evidence for the early emergence of piperaquine-resistant Plasmodium falciparum malaria and modeling strategies to mitigate resistance. PLoS Pathog 2022; 18:e1010278. [PMID: 35130315 PMCID: PMC8853508 DOI: 10.1371/journal.ppat.1010278] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/17/2022] [Accepted: 01/13/2022] [Indexed: 11/19/2022] Open
Abstract
Multidrug-resistant Plasmodium falciparum parasites have emerged in Cambodia and neighboring countries in Southeast Asia, compromising the efficacy of first-line antimalarial combinations. Dihydroartemisinin + piperaquine (PPQ) treatment failure rates have risen to as high as 50% in some areas in this region. For PPQ, resistance is driven primarily by a series of mutant alleles of the P. falciparum chloroquine resistance transporter (PfCRT). PPQ resistance was reported in China three decades earlier, but the molecular driver remained unknown. Herein, we identify a PPQ-resistant pfcrt allele (China C) from Yunnan Province, China, whose genotypic lineage is distinct from the PPQ-resistant pfcrt alleles currently observed in Cambodia. Combining gene editing and competitive growth assays, we report that PfCRT China C confers moderate PPQ resistance while re-sensitizing parasites to chloroquine (CQ) and incurring a fitness cost that manifests as a reduced rate of parasite growth. PPQ transport assays using purified PfCRT isoforms, combined with molecular dynamics simulations, highlight differences in drug transport kinetics and in this transporter’s central cavity conformation between China C and the current Southeast Asian PPQ-resistant isoforms. We also report a novel computational model that incorporates empirically determined fitness landscapes at varying drug concentrations, combined with antimalarial susceptibility profiles, mutation rates, and drug pharmacokinetics. Our simulations with PPQ-resistant or -sensitive parasite lines predict that a three-day regimen of PPQ combined with CQ can effectively clear infections and prevent the evolution of PfCRT variants. This work suggests that including CQ in combination therapies could be effective in suppressing the evolution of PfCRT-mediated multidrug resistance in regions where PPQ has lost efficacy. The recent emergence of Plasmodium falciparum parasite resistance to the antimalarial drug piperaquine (PPQ) has contributed to frequent treatment failures across Southeast Asia, originating in Cambodia. Here, we show that earlier reports of PPQ resistance in Yunnan Province, China could be explained by the unique China C variant of the P. falciparum chloroquine resistance transporter PfCRT. Gene-edited parasites show a loss of fitness and parasite resensitization to the chemically related former first-line antimalarial chloroquine, while acquiring PPQ resistance via drug efflux. Molecular features of drug resistance were examined using biochemical assays to measure mutant PfCRT-mediated drug transport and molecular dynamics simulations with the recently solved PfCRT structure to assess changes in the central drug-binding cavity. We also describe a new computational model that incorporates parasite mutation rates, fitness costs, antimalarial susceptibilities, and drug pharmacological profiles to predict how infections with parasite strains expressing distinct PfCRT variants can evolve and be selected in response to different drug pressures and regimens. Simulations predict that a three-day regimen of PPQ plus chloroquine would be fully effective at preventing recrudescence of drug-resistant infections.
Collapse
Affiliation(s)
- Jennifer L Small-Saunders
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Laura M Hagenah
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Kathryn J Wicht
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Satish K Dhingra
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Ioanna Deni
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Jonathan Kim
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, New York United States of America
| | - Jeremie Vendome
- Schrödinger, Inc., New York, New York, United States of America
| | - Eva Gil-Iturbe
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Paul D Roepe
- Department of Chemistry, Georgetown University, Washington, DC, United States of America
- Department of Biochemistry and Cellular and Molecular Biology, Georgetown University, Washington, DC, United States of America
| | - Monica Mehta
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Filippo Mancia
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, New York United States of America
| | - Matthias Quick
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York, United States of America
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, New York, United States of America
- Center for Molecular Recognition, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Margaret J Eppstein
- Vermont Complex Systems Center, University of Vermont, Burlington, Vermont, United States of America
- Department of Computer Science, University of Vermont, Burlington, Vermont, United States of America
- Translational Global Infectious Diseases Research Center, University of Vermont, Burlington, Vermont, United States of America
| | - David A Fidock
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, United States of America
| |
Collapse
|
33
|
Abstract
Proteasomes are compartmentalized, ATP-dependent, N-terminal nucleophile hydrolases that play essentials roles in intracellular protein turnover. They are present in all 3 kingdoms. Pharmacological inhibition of proteasomes is detrimental to cell viability. Proteasome inhibitor rugs revolutionize the treatment of multiple myeloma. Proteasomes in pathogenic microbes such as Mycobacterium tuberculosis (Mtb), Plasmodium falciparum (Pf), and other parasites and worms have been validated as therapeutic targets. Starting with Mtb proteasome, efforts in developing inhibitors selective for microbial proteasomes have made great progress lately. In this review, we describe the strategies and pharmacophores that have been used in developing proteasome inhibitors with potency and selectivity that spare human proteasomes and highlight the development of clinical proteasome inhibitor candidates for treatment of leishmaniasis and Chagas disease. Finally, we discuss the future challenges and therapeutical potentials of the microbial proteasome inhibitors.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, New York, United States of America
| | - Gang Lin
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
34
|
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
|
35
|
Huntsman EM, Cho RM, Kogan HV, McNamara-Bordewick NK, Tomko RJ, Snow JW. Proteasome Inhibition Is an Effective Treatment Strategy for Microsporidia Infection in Honey Bees. Biomolecules 2021; 11:1600. [PMID: 34827599 PMCID: PMC8615682 DOI: 10.3390/biom11111600] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/26/2021] [Accepted: 10/26/2021] [Indexed: 12/26/2022] Open
Abstract
The microsporidia Nosema ceranae is an obligate intracellular parasite that causes honey bee mortality and contributes to colony collapse. Fumagillin is presently the only pharmacological control for N. ceranae infections in honey bees. Resistance is already emerging, and alternative controls are critically needed. Nosema spp. exhibit increased sensitivity to heat shock, a common proteotoxic stress. Thus, we hypothesized that targeting the Nosema proteasome, the major protease removing misfolded proteins, might be effective against N. ceranae infections in honey bees. Nosema genome analysis and molecular modeling revealed an unexpectedly compact proteasome apparently lacking multiple canonical subunits, but with highly conserved proteolytic active sites expected to be receptive to FDA-approved proteasome inhibitors. Indeed, N. ceranae were strikingly sensitive to pharmacological disruption of proteasome function at doses that were well tolerated by honey bees. Thus, proteasome inhibition is a novel candidate treatment strategy for microsporidia infection in honey bees.
Collapse
Affiliation(s)
- Emily M. Huntsman
- Biology Department, Barnard College, New York, NY 10027, USA; (E.M.H.); (R.M.C.); (H.V.K.); (N.K.M.-B.)
| | - Rachel M. Cho
- Biology Department, Barnard College, New York, NY 10027, USA; (E.M.H.); (R.M.C.); (H.V.K.); (N.K.M.-B.)
| | - Helen V. Kogan
- Biology Department, Barnard College, New York, NY 10027, USA; (E.M.H.); (R.M.C.); (H.V.K.); (N.K.M.-B.)
| | | | - Robert J. Tomko
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA;
| | - Jonathan W. Snow
- Biology Department, Barnard College, New York, NY 10027, USA; (E.M.H.); (R.M.C.); (H.V.K.); (N.K.M.-B.)
| |
Collapse
|
36
|
Design of proteasome inhibitors with oral efficacy in vivo against Plasmodium falciparum and selectivity over the human proteasome. Proc Natl Acad Sci U S A 2021; 118:2107213118. [PMID: 34548400 PMCID: PMC8488693 DOI: 10.1073/pnas.2107213118] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2021] [Indexed: 11/18/2022] Open
Abstract
Here, we describe inhibitors of the Plasmodium proteasome, an enzymatic complex that malaria parasites rely on to degrade proteins. Starting from inhibitors developed to treat cancer, derivatives were designed and synthesized with the aim of increasing potency against the Plasmodium proteasome and decreasing activity against the human enzyme. Biochemical and cellular assays identified compounds that exhibit selectivity and potency, both in vitro and in vivo, at different stages of the parasite’s lifecycle. Cryo-electron microscopy revealed that the inhibitors bind in a hydrophobic pocket that is structurally different in the human proteasome—underpinning their selectivity. The work will help develop antimalarial therapeutics, which are desperately needed to treat a disease that kills nearly half a million people annually. The Plasmodium falciparum proteasome is a potential antimalarial drug target. We have identified a series of amino-amide boronates that are potent and specific inhibitors of the P. falciparum 20S proteasome (Pf20S) β5 active site and that exhibit fast-acting antimalarial activity. They selectively inhibit the growth of P. falciparum compared with a human cell line and exhibit high potency against field isolates of P. falciparum and Plasmodium vivax. They have a low propensity for development of resistance and possess liver stage and transmission-blocking activity. Exemplar compounds, MPI-5 and MPI-13, show potent activity against P. falciparum infections in a SCID mouse model with an oral dosing regimen that is well tolerated. We show that MPI-5 binds more strongly to Pf20S than to human constitutive 20S (Hs20Sc). Comparison of the cryo-electron microscopy (EM) structures of Pf20S and Hs20Sc in complex with MPI-5 and Pf20S in complex with the clinically used anti-cancer agent, bortezomib, reveal differences in binding modes that help to explain the selectivity. Together, this work provides insights into the 20S proteasome in P. falciparum, underpinning the design of potent and selective antimalarial proteasome inhibitors.
Collapse
|
37
|
Cheuka PM. Drug Discovery and Target Identification against Schistosomiasis: a Reality Check on Progress and Future Prospects. Curr Top Med Chem 2021; 22:1595-1610. [PMID: 34565320 DOI: 10.2174/1568026621666210924101805] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 11/22/2022]
Abstract
Schistosomiasis ranks among the most important infectious diseases, with over 200 million people currently being infected and > 280,000 deaths reported annually. Chemotherapeutic treatment has relied on one drug, praziquantel, for four decades, while other drugs, such as oxamniquine and metrifonate, are no longer preferred for clinical use due to their narrow spectrum of activity - these are only active against S. mansoni and S. haematobium, respectively. Despite being cheap, safe, and effective against all schistosome species, praziquantel is ineffective against immature worms, which may lead to reinfections and treatment failure in endemic areas; a situation that necessitates repeated administration besides other limitations. Therefore, novel drugs are urgently needed to overcome this situation. In this paper, an up to date review of drug targets identified and validated against schistosomiasis while also encompassing promising clinical and preclinical candidate drugs is presented. While there are considerable efforts aimed at identifying and validating drug targets, the pipeline for new antischistosomals is dry. Moreover, the majority of compounds evaluated preclinically are not really advanced because most of them were evaluated in very small preclinical species such as mice alone. Overall, it appears that although a lot of research is going on at discovery phases, unfortunately, it does not translate to advanced preclinical and clinical evaluation.
Collapse
Affiliation(s)
- Peter Mubanga Cheuka
- Department of Chemistry, School of Natural Sciences, University of Zambia, Lusaka. Zambia
| |
Collapse
|
38
|
Abstract
All living organisms depend on tightly regulated cellular networks to control biological functions. Proteolysis is an important irreversible post-translational modification that regulates most, if not all, cellular processes. Proteases are a large family of enzymes that perform hydrolysis of protein substrates, leading to protein activation or degradation. The 473 known and 90 putative human proteases are divided into 5 main mechanistic groups: metalloproteases, serine proteases, cysteine proteases, threonine proteases, and aspartic acid proteases. Proteases are fundamental to all biological systems, and when dysregulated they profoundly influence disease progression. Inhibiting proteases has led to effective therapies for viral infections, cardiovascular disorders, and blood coagulation just to name a few. Between 5 and 10% of all pharmaceutical targets are proteases, despite limited knowledge about their biological roles. More than 50% of all human proteases have no known substrates. We present here a comprehensive list of all current known human proteases. We also present current and novel biochemical tools to characterize protease functions in vitro, in vivo, and ex vivo. These tools make it achievable to define both beneficial and detrimental activities of proteases in health and disease.
Collapse
Affiliation(s)
- Longxiang Wang
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Kimberly Main
- Department of Physiology & Pharmacology, University of Calgary, Calgary, AB T2N 1N4, Canada.,McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB T2N 1N4, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Henry Wang
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Olivier Julien
- Department of Biochemistry, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Antoine Dufour
- Department of Physiology & Pharmacology, University of Calgary, Calgary, AB T2N 1N4, Canada.,McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, AB T2N 1N4, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
| |
Collapse
|
39
|
The apicoplast link to fever-survival and artemisinin-resistance in the malaria parasite. Nat Commun 2021; 12:4563. [PMID: 34315897 PMCID: PMC8316339 DOI: 10.1038/s41467-021-24814-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 07/09/2021] [Indexed: 01/02/2023] Open
Abstract
The emergence and spread of Plasmodium falciparum parasites resistant to front-line antimalarial artemisinin-combination therapies (ACT) threatens to erase the considerable gains against the disease of the last decade. Here, we develop a large-scale phenotypic screening pipeline and use it to carry out a large-scale forward-genetic phenotype screen in P. falciparum to identify genes allowing parasites to survive febrile temperatures. Screening identifies more than 200 P. falciparum mutants with differential responses to increased temperature. These mutants are more likely to be sensitive to artemisinin derivatives as well as to heightened oxidative stress. Major processes critical for P. falciparum tolerance to febrile temperatures and artemisinin include highly essential, conserved pathways associated with protein-folding, heat shock and proteasome-mediated degradation, and unexpectedly, isoprenoid biosynthesis, which originated from the ancestral genome of the parasite’s algal endosymbiont-derived plastid, the apicoplast. Apicoplast-targeted genes in general are upregulated in response to heat shock, as are other Plasmodium genes with orthologs in plant and algal genomes. Plasmodium falciparum parasites appear to exploit their innate febrile-response mechanisms to mediate resistance to artemisinin. Both responses depend on endosymbiont-derived genes in the parasite’s genome, suggesting a link to the evolutionary origins of Plasmodium parasites in free-living ancestors. Repeating fever is a hallmark of malaria. Here, a large-scale forward genetic screen in malaria-causing Plasmodium falciparum identifies genes associated with parasite tolerance to host fever, including apicoplast targeted isoprenoid biosynthesis—sharing features with artemisinin resistance.
Collapse
|
40
|
Behrens HM, Schmidt S, Spielmann T. The newly discovered role of endocytosis in artemisinin resistance. Med Res Rev 2021; 41:2998-3022. [PMID: 34309894 DOI: 10.1002/med.21848] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/15/2021] [Accepted: 07/03/2021] [Indexed: 12/28/2022]
Abstract
Artemisinin and its derivatives (ART) are the cornerstone of malaria treatment as part of artemisinin combination therapy (ACT). However, reduced susceptibility to artemisinin as well as its partner drugs threatens the usefulness of ACTs. Single point mutations in the parasite protein Kelch13 (K13) are necessary and sufficient for the reduced sensitivity of malaria parasites to ART but several alternative mechanisms for this resistance have been proposed. Recent work found that K13 is involved in the endocytosis of host cell cytosol and indicated that this is the process responsible for resistance in parasites with mutated K13. These studies also identified a series of further proteins that act together with K13 in the same pathway, including previously suspected resistance proteins such as UBP1 and AP-2μ. Here, we give a brief overview of artemisinin resistance, present the recent evidence of the role of endocytosis in ART resistance and discuss previous hypotheses in light of this new evidence. We also give an outlook on how the new insights might affect future research.
Collapse
Affiliation(s)
- Hannah Michaela Behrens
- Molecular Biology and Immunology Section, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Sabine Schmidt
- Molecular Biology and Immunology Section, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Tobias Spielmann
- Molecular Biology and Immunology Section, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| |
Collapse
|
41
|
Rosenthal MR, Ng CL. A Proteasome Mutation Sensitizes P. falciparum Cam3.II K13 C580Y Parasites to DHA and OZ439. ACS Infect Dis 2021; 7:1923-1931. [PMID: 33971094 PMCID: PMC8500539 DOI: 10.1021/acsinfecdis.0c00900] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Artemisinin-based combination therapies (ACTs), the World Health Organization-recommended first-line therapy for uncomplicated falciparum malaria, has led to significant decreases in malaria-associated morbidity and mortality in the past two decades. Decreased therapeutic efficacy of artemisinins, the cornerstone of ACTs, is threatening the gains made against this disease. As such, novel therapeutics with uncompromised mechanisms of action are needed to combat parasite-mediated antimalarial resistance. We have previously reported the antimalarial activity of Plasmodium falciparum-specific proteasome inhibitors in conjunction with a variety of antimalarials in clinical use or in preclinical investigations and of proteasome mutants generated in response to these inhibitors. Here, we discover that despite harboring K13C580Y, which has conventionally mediated artemisinin resistance in vitro as measured by increased survival in ring-stage survival assays (RSA), the Cam3.II strain parasites of Cambodian origin that have acquired an additional mutation in the proteasome display increased susceptibility to DHA and OZ439. This discovery implicates the proteasome in peroxide susceptibilities and has favorable implications on the use of peroxide and proteasome inhibitor combination therapy for the treatment of artemisinin-resistant malaria.
Collapse
Affiliation(s)
- Melissa R. Rosenthal
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Caroline L. Ng
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
42
|
Sharon M, Regev-Rudzki N. Cell communication and protein degradation: All in one parasitic package. J Extracell Vesicles 2021; 10:e12116. [PMID: 34257846 PMCID: PMC8256287 DOI: 10.1002/jev2.12116] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/18/2021] [Indexed: 01/12/2023] Open
Affiliation(s)
- Michal Sharon
- Department of Bimolecular Sciences Weizmann Institute of Science Rehovot Israel
| | - Neta Regev-Rudzki
- Department of Bimolecular Sciences Weizmann Institute of Science Rehovot Israel
| |
Collapse
|
43
|
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
|
44
|
Mata-Cantero L, Xie SC, García M, Coyle J, Fernandez R, Cabrera AC, Gillett DL, Crespo B, Gamo FJ, Fernández E, Tilley L, Gomez-Lorenzo MG. High Throughput Screening to Identify Selective and Nonpeptidomimetic Proteasome Inhibitors As Antimalarials. ACS Infect Dis 2021; 7:1818-1832. [PMID: 34044540 DOI: 10.1021/acsinfecdis.1c00110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The Ubiquitin Proteasome System is the main proteolytic pathway in eukaryotic cells, playing a role in key cellular processes. The essentiality of the Plasmodium falciparum proteasome is well validated, underlying its potential as an antimalarial target, but selective compounds are required to avoid cytotoxic effects in humans. Almost 550000 compounds were tested for the inhibition of the chymotrypsin-like activity of the P. falciparum proteasome using a Proteasome-GLO luminescence assay. Hits were confirmed in an orthogonal enzyme assay using Rho110-labeled peptides, and selectivity was assessed against the human proteasome. Four nonpeptidomimetic chemical families with some selectivity for the P. falciparum proteasome were identified and characterized in assays of proteasome trypsin and caspase activities and in parasite growth inhibition assays. Target engagement studies were performed, validating our approach. Hits identified are good starting points for the development of new antimalarial drugs and as tools to better understand proteasome function in P. falciparum.
Collapse
Affiliation(s)
- Lydia Mata-Cantero
- Global Health Discovery Incubator Unit, Global Health R&D. GlaxoSmithKline. Severo Ochoa 2, Tres Cantos 28760, Madrid Spain
| | - Stanley C. Xie
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, 30 Flemington Road, Melbourne, Victoria 3010, Australia
| | - Mercedes García
- MST-MedDesign-MedChem R&D. GlaxoSmithKline. Severo Ochoa 2, Tres Cantos 28760, Madrid Spain
| | - Joanne Coyle
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, 30 Flemington Road, Melbourne, Victoria 3010, Australia
| | - Raquel Fernandez
- MST-MedDesign-MedChem R&D. GlaxoSmithKline. Severo Ochoa 2, Tres Cantos 28760, Madrid Spain
| | - Alvaro Cortes Cabrera
- MST-MedDesign-MedChem R&D. GlaxoSmithKline. Severo Ochoa 2, Tres Cantos 28760, Madrid Spain
| | - David L. Gillett
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, 30 Flemington Road, Melbourne, Victoria 3010, Australia
| | - Benigno Crespo
- Global Health Discovery Incubator Unit, Global Health R&D. GlaxoSmithKline. Severo Ochoa 2, Tres Cantos 28760, Madrid Spain
| | - Francisco-Javier Gamo
- Global Health Discovery Incubator Unit, Global Health R&D. GlaxoSmithKline. Severo Ochoa 2, Tres Cantos 28760, Madrid Spain
| | - Esther Fernández
- Global Health Discovery Incubator Unit, Global Health R&D. GlaxoSmithKline. Severo Ochoa 2, Tres Cantos 28760, Madrid Spain
| | - Leann Tilley
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, 30 Flemington Road, Melbourne, Victoria 3010, Australia
| | - Maria G. Gomez-Lorenzo
- Global Health Discovery Incubator Unit, Global Health R&D. GlaxoSmithKline. Severo Ochoa 2, Tres Cantos 28760, Madrid Spain
| |
Collapse
|
45
|
Zhan W, Zhang H, Ginn J, Leung A, Liu YJ, Michino M, Toita A, Okamoto R, Wong TT, Imaeda T, Hara R, Yukawa T, Chelebieva S, Tumwebaze PK, Lafuente-Monasterio MJ, Martinez-Martinez MS, Vendome J, Beuming T, Sato K, Aso K, Rosenthal PJ, Cooper RA, Meinke PT, Nathan CF, Kirkman LA, Lin G. Development of a Highly Selective Plasmodium falciparum Proteasome Inhibitor with Anti-malaria Activity in Humanized Mice. Angew Chem Int Ed Engl 2021; 60:9279-9283. [PMID: 33433953 PMCID: PMC8087158 DOI: 10.1002/anie.202015845] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/29/2020] [Indexed: 01/01/2023]
Abstract
Plasmodium falciparum proteasome (Pf20S) inhibitors are active against Plasmodium at multiple stages-erythrocytic, gametocyte, liver, and gamete activation stages-indicating that selective Pf20S inhibitors possess the potential to be therapeutic, prophylactic, and transmission-blocking antimalarials. Starting from a reported compound, we developed a noncovalent, macrocyclic peptide inhibitor of the malarial proteasome with high species selectivity and improved pharmacokinetic properties. The compound demonstrates specific, time-dependent inhibition of the β5 subunit of the Pf20S, kills artemisinin-sensitive and artemisinin-resistant P. falciparum isolates in vitro and reduces parasitemia in humanized, P. falciparum-infected mice.
Collapse
Affiliation(s)
- Wenhu Zhan
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| | - 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
| | - Annie Leung
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| | - Yi J Liu
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, 1300 York Ave, New York, NY, 10065, USA
| | - Mayako Michino
- Tri-Institutional Therapeutics Discovery Institute, 413 E 69th St, 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
| | - 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
| | | | - Maria Jose Lafuente-Monasterio
- Diseases of the Developing World (DDW), Tres Cantos Medicine Development Campus, GlaxoSmithKline, Severo Ochoa 2, 28760, Tres Cantos, Madrid, Spain
| | - Maria Santos Martinez-Martinez
- Diseases of the Developing World (DDW), Tres Cantos Medicine Development Campus, GlaxoSmithKline, Severo Ochoa 2, 28760, Tres Cantos, Madrid, Spain
| | | | | | - 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
| | - 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
- Department of Medicine, Division of Infectious Diseases, 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
|
46
|
Zhan W, Zhang H, Ginn J, Leung A, Liu YJ, Michino M, Toita A, Okamoto R, Wong T, Imaeda T, Hara R, Yukawa T, Chelebieva S, Tumwebaze PK, Lafuente‐Monasterio MJ, Martinez‐Martinez MS, Vendome J, Beuming T, Sato K, Aso K, Rosenthal PJ, Cooper RA, Meinke PT, Nathan CF, Kirkman LA, Lin G. Development of a Highly Selective
Plasmodium falciparum
Proteasome Inhibitor with Anti‐malaria Activity in Humanized Mice. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202015845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Wenhu Zhan
- Department of Microbiology & Immunology Weill Cornell Medicine 1300 York Ave New York NY 10065 USA
| | - 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
| | - Annie Leung
- Department of Medicine Division of Infectious Diseases Weill Cornell Medicine 1300 York Ave New York NY 10065 USA
| | - Yi J. Liu
- Department of Medicine Division of Infectious Diseases Weill Cornell Medicine 1300 York Ave New York NY 10065 USA
| | - Mayako Michino
- Tri-Institutional Therapeutics Discovery Institute 413 E 69th St 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
| | - 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
| | | | - Maria Jose Lafuente‐Monasterio
- Diseases of the Developing World (DDW) Tres Cantos Medicine Development Campus GlaxoSmithKline Severo Ochoa 2 28760, Tres Cantos Madrid Spain
| | - Maria Santos Martinez‐Martinez
- Diseases of the Developing World (DDW) Tres Cantos Medicine Development Campus GlaxoSmithKline Severo Ochoa 2 28760, Tres Cantos Madrid Spain
| | | | | | - 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
| | | | - Roland A. Cooper
- Department of Natural Sciences and Mathematics Dominican University of California San Rafael CA 94901 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
- Department of Medicine Division of Infectious Diseases 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
|
47
|
Onoabedje EA, Ibezim A, Okoro UC, Batra S. New sulphonamide pyrolidine carboxamide derivatives: Synthesis, molecular docking, antiplasmodial and antioxidant activities. PLoS One 2021; 16:e0243305. [PMID: 33626047 PMCID: PMC7904193 DOI: 10.1371/journal.pone.0243305] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 11/18/2020] [Indexed: 11/22/2022] Open
Abstract
Carboxamides bearing sulphonamide functionality have been shown to exhibit significant lethal effect on Plasmodium falciparum, the causative agent of human malaria. Here we report the synthesis of thirty-two new drug-like sulphonamide pyrolidine carboxamide derivatives and their antiplasmodial and antioxidant capabilities. In addition, molecular docking was used to check their binding affinities for homology modelled P. falciparum N-myristoyltransferase, a confirmed drug target in the pathogen. Results revealed that sixteen new derivatives killed the parasite at single-digit micromolar concentration (IC50 = 2.40–8.30 μM) and compounds 10b, 10c, 10d, 10j and 10o scavenged DPPH radicals at IC50s (6.48, 8.49, 3.02, 6.44 and 4.32 μg/mL respectively) comparable with 1.06 μg/mL for ascorbic acid. Compound 10o emerged as the most active of the derivatives to bind to the PfNMT with theoretical inhibition constant (Ki = 0.09 μM) comparable to the reference ligand pyrazole-sulphonamide (Ki = 0.01 μM). This study identifies compound 10o, and this series in general, as potential antimalarial candidate with antioxidant activity which requires further attention to optimise activity.
Collapse
Affiliation(s)
- Efeturi A. Onoabedje
- Department of Pure & Industrial Chemistry, Faculty of Physical Sciences, University of Nigeria, Nsukka, Enugu State, Nigeria
- Division of Medicinal & Process Chemistry, Central Drug Research Institute, Lucknow, UP, India
- * E-mail: (EAO); (AI)
| | - Akachukwu Ibezim
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka, Enugu State, Nigeria
- * E-mail: (EAO); (AI)
| | - Uchechukwu C. Okoro
- Department of Pure & Industrial Chemistry, Faculty of Physical Sciences, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Sanjay Batra
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka, Enugu State, Nigeria
| |
Collapse
|
48
|
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
|
49
|
Zhan W, Singh PK, Ban Y, Qing X, Ah Kioon MD, Fan H, Zhao Q, Wang R, Sukenick G, Salmon J, Warren JD, Ma X, Barrat FJ, Nathan CF, Lin G. Structure-Activity Relationships of Noncovalent Immunoproteasome β5i-Selective Dipeptides. J Med Chem 2020; 63:13103-13123. [PMID: 33095579 PMCID: PMC8086754 DOI: 10.1021/acs.jmedchem.0c01520] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The immunoproteasome (i-20S) has emerged as a therapeutic target for autoimmune and inflammatory disorders and hematological malignancies. Inhibition of the chymotryptic β5i subunit of i-20S inhibits T cell activation, B cell proliferation, and dendritic cell differentiation in vitro and suppresses immune responses in animal models of autoimmune disorders and allograft rejection. However, cytotoxicity to immune cells has accompanied the use of covalently reactive β5i inhibitors, whose activity against the constitutive proteasome (c-20S) is cumulative with the time of exposure. Herein, we report a structure-activity relationship study of a class of noncovalent proteasome inhibitors with picomolar potencies and 1000-fold selectivity for i-20S over c-20S. Furthermore, these inhibitors are specific for β5i over the other five active subunits of i-20S and c-20S, providing useful tools to study the functions of β5i in immune responses. The potency of these compounds in inhibiting human T cell activation suggests that they may have therapeutic potential.
Collapse
Affiliation(s)
- Wenhu Zhan
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065
| | - Pradeep K Singh
- Department of Biochemistry, Milstein Chemistry Core Facility
| | - Yi Ban
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065
| | - Xiaoping Qing
- Autoimmunity and Inflammation Program, HSS Research Institute, Hospital for Special Surgery, New York, NY 10065, USA
| | - Marie Dominique Ah Kioon
- Autoimmunity and Inflammation Program, HSS Research Institute, Hospital for Special Surgery, New York, NY 10065, USA
| | - Hao Fan
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065
| | - Quanju Zhao
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065
| | - Rong Wang
- NMR Analytical Core Facility, Memorial Sloan Kettering Cancer Center
| | - George Sukenick
- NMR Analytical Core Facility, Memorial Sloan Kettering Cancer Center
| | - Jane Salmon
- Autoimmunity and Inflammation Program, HSS Research Institute, Hospital for Special Surgery, New York, NY 10065, USA
| | - J David Warren
- Department of Biochemistry, Milstein Chemistry Core Facility
| | - Xiaojing Ma
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065
| | - Franck J. Barrat
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065
- Autoimmunity and Inflammation Program, HSS Research Institute, Hospital for Special Surgery, New York, NY 10065, USA
| | - Carl F. Nathan
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065
| | - Gang Lin
- Department of Microbiology & Immunology, Weill Cornell Medicine, 1300 York Ave, New York, NY 10065
| |
Collapse
|
50
|
Yang J, He Y, Li Y, Zhang X, Wong YK, Shen S, Zhong T, Zhang J, Liu Q, Wang J. Advances in the research on the targets of anti-malaria actions of artemisinin. Pharmacol Ther 2020; 216:107697. [PMID: 33035577 PMCID: PMC7537645 DOI: 10.1016/j.pharmthera.2020.107697] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/30/2020] [Accepted: 09/30/2020] [Indexed: 01/01/2023]
Abstract
Malaria has been a global epidemic health threat since ancient times. It still claims roughly half a million lives every year in this century. Artemisinin and its derivatives, are frontline antimalarial drugs known for their efficacy and low toxicity. After decades of wide use, artemisinins remain our bulwark against malaria. Here, we review decades of efforts that aim to understand the mechanism of action (MOA) of artemisinins, which help explain the specificity and potency of this anti-malarial drug. We summarize the methods and approaches employed to unravel the MOA of artemisinin over the last three decades, showing how the development of advanced techniques can help provide mechanistic insights and resolve some long-standing questions in the field of artemisinin research. We also provide examples to illustrate how to better repurpose artemisinins for anti-cancer therapies by leveraging on MOA. These examples point out a practical direction to engineer artemisinin for broader applications beyond malaria.
Collapse
Affiliation(s)
- Jing Yang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China; Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yingke He
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China; Department of Anaesthesiology, Singapore General Hospital, Singapore
| | - Yinbao Li
- School of Pharmaceutical Sciences, Gannan Medical University, Ganzhou, JiangXi 341000, China
| | - Xing Zhang
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yin-Kwan Wong
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shengnan Shen
- Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tianyu Zhong
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China; Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China.
| | - Jianbin Zhang
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.
| | - Qian Liu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China.
| | - Jigang Wang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China; Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China; Central People's Hospital of Zhanjiang, Zhanjiang, Guangdong, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|