1
|
de Oliveira Rios É, Albino SL, Olimpio de Moura R, Nascimento IJDS. Targeting cysteine protease B to discover antileishmanial drugs: Directions and advances. Eur J Med Chem 2025; 289:117500. [PMID: 40085977 DOI: 10.1016/j.ejmech.2025.117500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/27/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Leishmaniasis is a severe disease and results in high mortality rates. Despite this, there are few drugs to treat and with various limitations such as toxicity and resistance, which justifies the search for new drugs. Thus, cysteine protease B (CPB) is a promising target against leishmania due to its immunomodulatory function related to the parasite's virulence and its interaction with the host. Thus, this perspective showed the potential of CPB in drug design and the main insights that can be used in subsequent drug design works. In fact, the aziridine analogs are the most explored against CPB due to the promising results and provide several insights into drug design. Also, it is noteworthy that one of the biggest challenges is target selectivity. Knowledge about substrate binding and other factors, such as the reversibility of inhibitors, is also needed. In addition, exploring target selectivity patterns is critical to developing CP inhibitors for clinical use to combat this threatening agent.
Collapse
Affiliation(s)
| | - Sonaly Lima Albino
- Postgraduate Program of Pharmaceutical Sciences, Pharmacy Department, State University of Paraíba, Campina Grande, PB, Brazil; Laboratory of Synthesis and Drug Delivery, Department of Biological Sciences, State University of Paraiba, João Pessoa, Brazil
| | - Ricardo Olimpio de Moura
- Postgraduate Program of Pharmaceutical Sciences, Pharmacy Department, State University of Paraíba, Campina Grande, PB, Brazil; Laboratory of Synthesis and Drug Delivery, Department of Biological Sciences, State University of Paraiba, João Pessoa, Brazil
| | - Igor José Dos Santos Nascimento
- Cesmac University Center, Pharmacy Department, Maceió, Brazil; Postgraduate Program of Pharmaceutical Sciences, Pharmacy Department, State University of Paraíba, Campina Grande, PB, Brazil; Laboratory of Synthesis and Drug Delivery, Department of Biological Sciences, State University of Paraiba, João Pessoa, Brazil.
| |
Collapse
|
2
|
Sanaullah B, Truong NV, Nguyen TK, Han ET. Combating Malaria: Targeting the Ubiquitin-Proteasome System to Conquer Drug Resistance. Trop Med Infect Dis 2025; 10:94. [PMID: 40278767 PMCID: PMC12031434 DOI: 10.3390/tropicalmed10040094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
Malaria primarily affects developing nations and is one of the most destructive and pervasive tropical parasite infections. Antimalarial drug resistance, characterized by a parasite's ability to survive and reproduce despite recommended medication doses, poses a significant challenge. Along with resistance to antimalarial drugs, the rate of mutation a parasite undergoes, overall parasite load, drug potency, adherence to treatment, dosing accuracy, drug bioavailability, and the presence of poor-quality counterfeit drugs are some of the contributing factors that elicit opposition to treatment. The ubiquitin-proteasome system (UPS) has become a promising drug target for malaria because of its central importance in the parasite's life cycle and its contribution to artemisinin resistance. Polymorphisms in the Kelch13 gene of Plasmodium falciparum are the best-known markers for artemisinin resistance and are associated with a highly active UPS. Certain proteasome inhibitors, which are the other key players of the UPS, have demonstrated activity against malarial parasites and the ability to work with artemisinin. This work describes how, through targeting the UPS, the greater effectiveness of antimalarial drugs-especially where there is strong resistance-can be achieved, which contributes to overcoming the drug resistance phenomenon in malaria.
Collapse
Affiliation(s)
| | | | | | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
3
|
Okombo J, Fidock DA. Towards next-generation treatment options to combat Plasmodium falciparum malaria. Nat Rev Microbiol 2025; 23:178-191. [PMID: 39367132 PMCID: PMC11832322 DOI: 10.1038/s41579-024-01099-x] [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] [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
|
4
|
Yasir M, Park J, Han ET, Park WS, Han JH, Chun W. Structural comparison of human and Plasmodium proteasome β5 subunits: informing selective inhibitor design for anti-malaria agents. Malar J 2025; 24:21. [PMID: 39838389 PMCID: PMC11752634 DOI: 10.1186/s12936-025-05259-z] [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: 06/04/2024] [Accepted: 01/15/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND The Plasmodium proteasome emerges as a promising target for anti-malarial drug development due to its potential activity against multiple life cycle stages. METHODS In this investigation, a comparative analysis was conducted on the structural features of the β5 subunit in the 20S proteasomes of both Plasmodium and humans. RESULTS The findings underscore the structural diversity inherent in both proteasomes. The human proteasome β5 subunit reveals a composition rich in β-sheets and adopts a more compact conformation. This structural arrangement limits the ligand binding pocket's capacity to accommodate only small compounds effectively. In contrast, the Plasmodium β5 subunit exhibits a higher prevalence of loop structures, creating a more open and flexible binding pocket. This unique structural characteristic enables the binding of a larger and more diverse array of compounds. CONCLUSION The discernible structural contrast between the human and Plasmodium proteasome β5 subunits holds promise for the identification of Plasmodium-selective compounds. The ability of the Plasmodium proteasome to accommodate a broader range of compounds due to its distinctive structural features opens avenues for drug screening to intending to develop selective anti-malarial agents. This study contributes valuable insights into the structural basis for targeting the Plasmodium proteasome and paves the way for the rational design of compounds with enhanced specificity and efficacy against malaria.
Collapse
Affiliation(s)
- Muhammad Yasir
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, 24341, Republic of Korea
| | - Jinyoung Park
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, 24341, Republic of Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, 24341, Republic of Korea
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, Republic of Korea
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, 24341, Republic of Korea
| | - Wanjoo Chun
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
5
|
Eadsforth TC, Torrie LS, Rowland P, Edgar EV, MacLean LM, Paterson C, Robinson DA, Shepherd SM, Thomas J, Thomas MG, Gray DW, Postis VLG, De Rycker M. Pharmacological and structural understanding of the Trypanosoma cruzi proteasome provides key insights for developing site-specific inhibitors. J Biol Chem 2025; 301:108049. [PMID: 39638245 PMCID: PMC11748689 DOI: 10.1016/j.jbc.2024.108049] [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: 05/22/2024] [Revised: 11/15/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
The proteasome is considered an excellent drug target for many infectious diseases as well as cancer. Challenges with robust and safe supply of proteasomes from infectious agents, lack of structural information, and complex pharmacology due to multiple active sites have hampered progress in the infectious disease space. We recombinantly expressed the proteasome of the protozoan parasite Trypanosoma cruzi, the causative agent of Chagas disease, and demonstrate pharmacological equivalence to the native T. cruzi proteasome. Active-site mutant recombinant proteasomes reveal substrate promiscuity for WT proteasomes, with important implications for assessing pharmacological responses of active-site selective inhibitors. Using these mutant proteasomes, we show that some selective parasite proteasome inhibitors only partially inhibit the chymotrypsin-like activity, including a newly developed 5-(phenoxymethyl)furan-2-carboxamide-based proteasome inhibitor. In spite of partial inhibition, these compounds remain potent inhibitors of intracellular T. cruzi growth. Drug-resistant mutants provide further insights in drug mode-of-inhibition. We also present the high-resolution CryoEM structures of both native and recombinantly-expressed T. cruzi proteasomes which reveal pharmacologically relevant differences in the ligand-binding site compared to the related Leishmania proteasome. Furthermore, we show that the trypanosomatid β4/β5 selectivity pocket is not present in the proteasome structures of other protozoan parasites. This work highlights the need, and provides approaches, to precisely assess proteasome substrate selectivity and pharmacology. It enables structure-guided drug discovery for this promising Chagas disease drug target, provides a new chemical starting point for drug discovery, and paves the road for development of robust proteasome drug discovery programmes for other eukaryotic infectious diseases.
Collapse
Affiliation(s)
- Thomas C Eadsforth
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Leah S Torrie
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | | | | | - Lorna M MacLean
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Christy Paterson
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - David A Robinson
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Sharon M Shepherd
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - John Thomas
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Michael G Thomas
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - David W Gray
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Vincent L G Postis
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Manu De Rycker
- Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK.
| |
Collapse
|
6
|
Fajtova P, Hurysz BM, Miyamoto Y, Serafim MSM, Jiang Z, Vazquez JM, Trujillo DF, Liu LJ, Somani U, Almaliti J, Myers SA, Caffrey CR, Gerwick WH, McMinn DL, Kirk CJ, Boura E, Eckmann L, O'Donoghue AJ. Distinct substrate specificities of the three catalytic subunits of the Trichomonas vaginalis proteasome. Protein Sci 2024; 33:e5225. [PMID: 39589076 PMCID: PMC11590128 DOI: 10.1002/pro.5225] [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: 06/10/2024] [Revised: 09/25/2024] [Accepted: 11/06/2024] [Indexed: 11/27/2024]
Abstract
The protozoan parasite Trichomonas vaginalis (Tv) causes trichomoniasis, the most common non-viral sexually transmitted infection in the world. Although Tv has been linked to significant health complications, only two closely related 5-nitroimidazole drugs are approved for its treatment. The emergence of resistance to these drugs and lack of alternative treatment options poses an increasing threat to public health, making development of novel anti-Trichomonas compounds an urgent need. The proteasome, a critical enzyme complex found in all eukaryotes has three catalytic subunits, β1, β2, and β5 and has been validated as a drug target to treat trichomoniasis. With the goal of developing tools to study the Tv proteasome, we isolated the enzyme complex and identified inhibitors that preferentially inactivate either one or two of the three catalytic subunits. Using a mass spectrometry-based peptide digestion assay, these inhibitors were used to define the substrate preferences of the β1, β2 and β5 subunits. Subsequently, three model fluorogenic substrates were designed, each specific for one of the catalytic subunits. This novel substrate profiling methodology will allow for individual subunit characterization of other proteasomes of interest. Using the new substrates, we screened a library of 284 peptide epoxyketone inhibitors against Tv and determined the subunits targeted by the most active compounds. The data show that inhibition of the Tv β5 subunit alone is toxic to the parasite. Taken together, the optimized proteasome subunit substrates will be instrumental for understanding the molecular determinants of proteasome specificity and for accelerating drug development against trichomoniasis.
Collapse
Affiliation(s)
- Pavla Fajtova
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California San DiegoLa JollaCaliforniaUSA
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of SciencesPrague 6Czech Republic
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Brianna M. Hurysz
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California San DiegoLa JollaCaliforniaUSA
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Yukiko Miyamoto
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaLa JollaCaliforniaUSA
- Department of MedicineUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Mateus Sá M. Serafim
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California San DiegoLa JollaCaliforniaUSA
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaLa JollaCaliforniaUSA
- Departamento de Microbiologia, Instituto de Ciências BiológicasUniversidade Federal de Minas Gerais (UFMG)Belo HorizonteMinas GeraisBrazil
| | - Zhenze Jiang
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California San DiegoLa JollaCaliforniaUSA
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Julia M. Vazquez
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California San DiegoLa JollaCaliforniaUSA
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Diego F. Trujillo
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California San DiegoLa JollaCaliforniaUSA
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Lawrence J. Liu
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California San DiegoLa JollaCaliforniaUSA
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Urvashi Somani
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California San DiegoLa JollaCaliforniaUSA
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | - Jehad Almaliti
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaLa JollaCaliforniaUSA
- Scripps Institution of OceanographyUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Samuel A. Myers
- Division of Signaling and Gene ExpressionLa Jolla Institute for ImmunologyLa JollaCaliforniaUSA
| | - Conor R. Caffrey
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California San DiegoLa JollaCaliforniaUSA
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| | - William H. Gerwick
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California San DiegoLa JollaCaliforniaUSA
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaLa JollaCaliforniaUSA
- Scripps Institution of OceanographyUniversity of California San DiegoLa JollaCaliforniaUSA
| | | | | | - Evzen Boura
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of SciencesPrague 6Czech Republic
| | - Lars Eckmann
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaLa JollaCaliforniaUSA
- Department of MedicineUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Anthony J. O'Donoghue
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California San DiegoLa JollaCaliforniaUSA
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of CaliforniaLa JollaCaliforniaUSA
| |
Collapse
|
7
|
Rosenthal MR, Vijayrajratnam S, Firestone TM, Ng CL. Enhanced cell stress response and protein degradation capacity underlie artemisinin resistance in Plasmodium falciparum. mSphere 2024; 9:e0037124. [PMID: 39436072 PMCID: PMC11580438 DOI: 10.1128/msphere.00371-24] [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: 05/03/2024] [Accepted: 09/03/2024] [Indexed: 10/23/2024] Open
Abstract
Malaria remains a global health burden, killing over half a million people each year. Decreased therapeutic efficacy to artemisinin, the most efficacious antimalarial, has been detected in sub-Saharan Africa, a worrying fact given that over 90% of deaths occur on this continent. Mutations in Kelch13 are the most well-established molecular marker for artemisinin resistance, but these do not explain all artemisinin-resistant isolates. Understanding the biological underpinnings of drug resistance is key to curbing the emergence and spread of artemisinin resistance. Artemisinin-mediated non-specific alkylation leads to the accumulation of misfolded and damaged proteins and activation of the parasite unfolded protein response (UPR). In addition, the parasite proteasome is vital to artemisinin resistance, as we have previously shown that chemical inhibition of the proteasome or mutations in the β2 proteasome subunit increase parasite susceptibility to dihydroartemisinin (DHA), the active metabolite of artemisinins. Here, we investigate parasites with mutations at the Kelch13 and/or 19S and 20S proteasome subunits with regard to UPR regulation and proteasome activity in the context of artemisinin resistance. Our data show that perturbing parasite proteostasis kills parasites, early parasite UPR signaling dictates DHA survival outcomes, and DHA susceptibility correlates with impairment of proteasome-mediated protein degradation. Importantly, we show that functional proteasomes are required for artemisinin resistance in a Kelch13-independent manner, and compound-selective proteasome inhibition demonstrates why artemisinin-resistant Kelch13 mutants remain susceptible to the related antimalarial peroxide OZ439. These data provide further evidence for targeting the parasite proteasome and UPR to overcome existing artemisinin resistance.IMPORTANCEDecreased therapeutic efficacy represents a major barrier to malaria treatment control strategies. The malaria proteasome and accompanying unfolded protein response are crucial to artemisinin resistance, revealing novel antimalarial therapeutic strategies.
Collapse
Affiliation(s)
- Melissa R. Rosenthal
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Sukhithasri Vijayrajratnam
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Tessa M. Firestone
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Caroline L. Ng
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Global Center for Health Security, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Biology, University of Omaha, Omaha, Nebraska, USA
| |
Collapse
|
8
|
Silhan J, Fajtova P, Bartosova J, Hurysz BM, Almaliti J, Miyamoto Y, Eckmann L, Gerwick WH, O'Donoghue AJ, Boura E. Structural elucidation of recombinant Trichomonas vaginalis 20S proteasome bound to covalent inhibitors. Nat Commun 2024; 15:8621. [PMID: 39366995 PMCID: PMC11452676 DOI: 10.1038/s41467-024-53022-w] [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: 09/22/2023] [Accepted: 09/27/2024] [Indexed: 10/06/2024] Open
Abstract
The proteasome is a proteolytic enzyme complex essential for protein homeostasis in mammalian cells and protozoan parasites like Trichomonas vaginalis (Tv), the cause of the most common, non-viral sexually transmitted disease. Tv and other protozoan 20S proteasomes have been validated as druggable targets for antimicrobials. However, low yields and purity of the native proteasome have hindered studies of the Tv 20S proteasome (Tv20S). We address this challenge by creating a recombinant protozoan proteasome by expressing all seven α and seven β subunits of Tv20S alongside the Ump-1 chaperone in insect cells. The recombinant Tv20S displays biochemical equivalence to its native counterpart, confirmed by various assays. Notably, the marizomib (MZB) inhibits all catalytic subunits of Tv20S, while the peptide inhibitor carmaphycin-17 (CP-17) specifically targets β2 and β5. Cryo-electron microscopy (cryo-EM) unveils the structures of Tv20S bound to MZB and CP-17 at 2.8 Å. These findings explain MZB's low specificity for Tv20S compared to the human proteasome and demonstrate CP-17's higher specificity. Overall, these data provide a structure-based strategy for the development of specific Tv20S inhibitors to treat trichomoniasis.
Collapse
Affiliation(s)
- Jan Silhan
- Institute of Organic Chemistry and Biochemistry AS CR, v.v.i., Prague, Czech Republic
| | - Pavla Fajtova
- Institute of Organic Chemistry and Biochemistry AS CR, v.v.i., Prague, Czech Republic.
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.
| | - Jitka Bartosova
- Institute of Organic Chemistry and Biochemistry AS CR, v.v.i., Prague, Czech Republic
| | - Brianna M Hurysz
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Jehad Almaliti
- Department Pharmaceutical Sciences, College of Pharmacy, The University of Jordan, Amman, Jordan
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, USA
| | - Yukiko Miyamoto
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Lars Eckmann
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - William H Gerwick
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, USA
| | - Anthony J O'Donoghue
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.
| | - Evzen Boura
- Institute of Organic Chemistry and Biochemistry AS CR, v.v.i., Prague, Czech Republic.
| |
Collapse
|
9
|
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
|
10
|
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 PMCID: PMC11531662 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
|
11
|
Joshi N, Hosen SKY, Fahad M, Narooka AR, Gourinath S, Tiwari S. The 26 S proteasome in Entamoeba histolytica: divergence of the substrate binding pockets from host proteasomes. BMC Res Notes 2024; 17:216. [PMID: 39095914 PMCID: PMC11295364 DOI: 10.1186/s13104-024-06848-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 06/26/2024] [Indexed: 08/04/2024] Open
Abstract
OBJECTIVE Proteasomes are conserved proteases crucial for proteostasis in eukaryotes and are promising drug targets for protozoan parasites. Yet, the proteasomes of Entamoeba histolytica remain understudied. The study's objective was to analyse the differences in the substrate binding pockets of amoeba proteasomes from those of host, and computational modelling of β5 catalytic subunit, with the goal of finding selective inhibitors. RESULTS Comparative sequence analysis revealed differences in substrate binding sites of E. histolytica proteasomes, especially in the S1 and S3 pockets of the catalytic beta subunits, implying differences in substrate preference and susceptibility to inhibitors from host proteasomes. This was strongly supported by significantly lower sensitivity to MG132 mediated inhibition of amoebic proteasome β5 subunit's chymotryptic activity compared to human proteasomes, also reflected in lower sensitivity of E. histolytica to MG132 for inhibition of proliferation. Computational models of β4 and β5 subunits, and a docked β4-β5 model revealed a binding pocket between β4-β5, similar to that of Leishmania tarentolae. Selective inhibitors for visceral leishmaniasis, LXE408 and compound 8, docked well to this pocket. This functional and sequence-based analysis predicts differences between amoebic and host proteasomes that can be utilized to develop rationally designed, selective inhibitors against E. histolytica.
Collapse
Affiliation(s)
- Nidhi Joshi
- Department of Pharmacology, University of Minnesota, Minneapolis, USA
- Molecular Cell Biology Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - S K Yasir Hosen
- Tata Institute of Fundamental Research, Hyderabad, 500046, India
- Molecular Cell Biology Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Mohd Fahad
- Molecular Cell Biology Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Anil Raj Narooka
- Proteomics Department, Advanced Enzymes Technologies Ltd, Thane, 400604, India
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
- Molecular Cell Biology Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - S Gourinath
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Swati Tiwari
- Molecular Cell Biology Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
12
|
Mansfield CR, Quan B, Chirgwin ME, Eduful B, Hughes PF, Neveu G, Sylvester K, Ryan DH, Kafsack BFC, Haystead TAJ, Leahy JW, Fitzgerald MC, Derbyshire ER. Selective targeting of Plasmodium falciparum Hsp90 disrupts the 26S proteasome. Cell Chem Biol 2024; 31:729-742.e13. [PMID: 38492573 PMCID: PMC11031320 DOI: 10.1016/j.chembiol.2024.02.008] [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: 03/01/2023] [Revised: 11/09/2023] [Accepted: 02/22/2024] [Indexed: 03/18/2024]
Abstract
The molecular chaperone heat shock protein 90 (Hsp90) has an essential but largely undefined role in maintaining proteostasis in Plasmodium falciparum, the most lethal malaria parasite. Herein, we identify BX-2819 and XL888 as potent P. falciparum (Pf)Hsp90 inhibitors. Derivatization of XL888's scaffold led to the development of Tropane 1, as a PfHsp90-selective binder with nanomolar affinity. Hsp90 inhibitors exhibit anti-Plasmodium activity against the liver, asexual blood, and early gametocyte life stages. Thermal proteome profiling was implemented to assess PfHsp90-dependent proteome stability, and the proteasome-the main site of cellular protein recycling-was enriched among proteins with perturbed stability upon PfHsp90 inhibition. Subsequent biochemical and cellular studies suggest that PfHsp90 directly promotes proteasome hydrolysis by chaperoning the active 26S complex. These findings expand our knowledge of the PfHsp90-dependent proteome and protein quality control mechanisms in these pathogenic parasites, as well as further characterize this chaperone as a potential antimalarial drug target.
Collapse
Affiliation(s)
- Christopher R Mansfield
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| | - Baiyi Quan
- Department of Chemistry, Duke University, Durham, NC, USA
| | | | - Benjamin Eduful
- Department of Chemistry, University of South Florida, Tampa, FL, USA
| | - Philip F Hughes
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Gaëlle Neveu
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Kayla Sylvester
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| | - Daniel H Ryan
- Department of Chemistry, Duke University, Durham, NC, USA
| | - Björn F C Kafsack
- Department of Microbiology & Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Timothy A J Haystead
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - James W Leahy
- Department of Chemistry, University of South Florida, Tampa, FL, USA; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; Center for Drug Discovery and Innovation, University of South Florida, Tampa, FL, USA
| | | | - Emily R Derbyshire
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA; Department of Chemistry, Duke University, Durham, NC, USA.
| |
Collapse
|
13
|
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
|
14
|
Pérez-Pertejo Y, García-Estrada C, Martínez-Valladares M, Murugesan S, Reguera RM, Balaña-Fouce R. Polyamine Metabolism for Drug Intervention in Trypanosomatids. Pathogens 2024; 13:79. [PMID: 38251386 PMCID: PMC10820115 DOI: 10.3390/pathogens13010079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
Neglected tropical diseases transmitted by trypanosomatids include three major human scourges that globally affect the world's poorest people: African trypanosomiasis or sleeping sickness, American trypanosomiasis or Chagas disease and different types of leishmaniasis. Different metabolic pathways have been targeted to find antitrypanosomatid drugs, including polyamine metabolism. Since their discovery, the naturally occurring polyamines, putrescine, spermidine and spermine, have been considered important metabolites involved in cell growth. With a complex metabolism involving biosynthesis, catabolism and interconversion, the synthesis of putrescine and spermidine was targeted by thousands of compounds in an effort to produce cell growth blockade in tumor and infectious processes with limited success. However, the discovery of eflornithine (DFMO) as a curative drug against sleeping sickness encouraged researchers to develop new molecules against these diseases. Polyamine synthesis inhibitors have also provided insight into the peculiarities of this pathway between the host and the parasite, and also among different trypanosomatid species, thus allowing the search for new specific chemical entities aimed to treat these diseases and leading to the investigation of target-based scaffolds. The main molecular targets include the enzymes involved in polyamine biosynthesis (ornithine decarboxylase, S-adenosylmethionine decarboxylase and spermidine synthase), enzymes participating in their uptake from the environment, and the enzymes involved in the redox balance of the parasite. In this review, we summarize the research behind polyamine-based treatments, the current trends, and the main challenges in this field.
Collapse
Affiliation(s)
- Yolanda Pérez-Pertejo
- Departamento de Ciencias Biomédicas, Campus de Vegazana s/n, Universidad de León, 24071 León, Spain; (Y.P.-P.); (C.G.-E.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Campus de Vegazana s/n, Universidad de León, 24071 León, Spain
| | - Carlos García-Estrada
- Departamento de Ciencias Biomédicas, Campus de Vegazana s/n, Universidad de León, 24071 León, Spain; (Y.P.-P.); (C.G.-E.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Campus de Vegazana s/n, Universidad de León, 24071 León, Spain
| | | | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani 333031, India;
| | - Rosa M. Reguera
- Departamento de Ciencias Biomédicas, Campus de Vegazana s/n, Universidad de León, 24071 León, Spain; (Y.P.-P.); (C.G.-E.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Campus de Vegazana s/n, Universidad de León, 24071 León, Spain
| | - Rafael Balaña-Fouce
- Departamento de Ciencias Biomédicas, Campus de Vegazana s/n, Universidad de León, 24071 León, Spain; (Y.P.-P.); (C.G.-E.); (R.M.R.)
- Instituto de Biomedicina (IBIOMED), Campus de Vegazana s/n, Universidad de León, 24071 León, Spain
| |
Collapse
|
15
|
Rostamighadi M, Kamelshahroudi A, Mehta V, Zeng FY, Pass I, Chung TDY, Salavati R. High-throughput screening of compounds targeting RNA editing in Trypanosoma brucei: Novel molecular scaffolds with broad trypanocidal effects. Biochem Pharmacol 2024; 219:115937. [PMID: 37995979 DOI: 10.1016/j.bcp.2023.115937] [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: 09/11/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023]
Abstract
Mitochondrial uridine insertion/deletion RNA editing, catalyzed by a multiprotein complex (editosome), is essential for gene expression in trypanosomes and Leishmania parasites. As this process is absent in the human host, a drug targeting this mechanism promises high selectivity and reduced toxicity. Here, we successfully miniaturized our FRET-based full-round RNA editing assay, which replicates the complete RNA editing process, adapting it into a 1536-well format. Leveraging this assay, we screened over 100,000 compounds against purified editosomes derived from Trypanosoma brucei, identifying seven confirmed primary hits. We sourced and evaluated various analogs to enhance the inhibitory and parasiticidal effects of these primary hits. In combination with secondary assays, our compounds marked inhibition of essential catalytic activities, including the RNA editing ligase and interactions of editosome proteins. Although the primary hits did not exhibit any growth inhibitory effect on parasites, we describe eight analog compounds capable of effectively killing T. brucei and/or Leishmania donovani parasites within a low micromolar concentration. Whether parasite killing is - at least in part - due to inhibition of RNA editing in vivo remains to be assessed. Our findings introduce novel molecular scaffolds with the potential for broad antitrypanosomal effects.
Collapse
Affiliation(s)
- Mojtaba Rostamighadi
- Institute of Parasitology, McGill University, Ste. Anne de Bellevue, Quebec H9X 3V9, Canada
| | - Arezou Kamelshahroudi
- Institute of Parasitology, McGill University, Ste. Anne de Bellevue, Quebec H9X 3V9, Canada
| | - Vaibhav Mehta
- Institute of Parasitology, McGill University, Ste. Anne de Bellevue, Quebec H9X 3V9, Canada; Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Fu-Yue Zeng
- Sanford Burnham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, La Jolla, CA, USA
| | - Ian Pass
- Sanford Burnham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, La Jolla, CA, USA
| | - Thomas D Y Chung
- Sanford Burnham Prebys Medical Discovery Institute, Conrad Prebys Center for Chemical Genomics, La Jolla, CA, USA
| | - Reza Salavati
- Institute of Parasitology, McGill University, Ste. Anne de Bellevue, Quebec H9X 3V9, Canada; Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada.
| |
Collapse
|
16
|
Silhan J, Fajtova P, Bartosova J, Hurysz BM, Almaliti J, Miyamoto Y, Eckmann L, Gerwick WH, O’Donoghue AJ, Boura E. Structural elucidation of recombinant Trichomonas vaginalis 20S proteasome bound to covalent inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.17.553660. [PMID: 37645851 PMCID: PMC10462138 DOI: 10.1101/2023.08.17.553660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Proteasomes are essential for protein homeostasis in mammalian cells1-4 and in protozoan parasites such as Trichomonas vaginalis (Tv).5 Tv and other protozoan 20S proteasomes have been validated as druggable targets.6-8 However, in the case of Tv 20S proteasome (Tv20S), biochemical and structural studies were impeded by low yields and purity of the native proteasome. We successfully made recombinant Tv20S by expressing all seven α and seven β subunits together with the Ump-1 chaperone in insect cells. We isolated recombinant proteasome and showed that it was biochemically indistinguishable from the native enzyme. We confirmed that the recombinant Tv20S is inhibited by the natural product marizomib (MZB)9 and the recently developed peptide inhibitor carmaphycin-17 (CP-17)8,10. Specifically, MZB binds to the β1, β2 and β5 subunits, while CP-17 binds the β2 and β5 subunits. Next, we obtained cryo-EM structures of Tv20S in complex with these covalent inhibitors at 2.8Å resolution. The structures revealed the overall fold of the Tv20S and the binding mode of MZB and CP-17. Our work explains the low specificity of MZB and higher specificity of CP-17 towards Tv20S as compared to human proteasome and provides the platform for the development of Tv20S inhibitors for treatment of trichomoniasis.
Collapse
Affiliation(s)
- Jan Silhan
- Institute of Organic Chemistry and Biochemistry AS CR, v.v.i., Flemingovo nam. 2., 166 10 Prague 6, Czech Republic
| | - Pavla Fajtova
- Institute of Organic Chemistry and Biochemistry AS CR, v.v.i., Flemingovo nam. 2., 166 10 Prague 6, Czech Republic
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92037, USA
| | - Jitka Bartosova
- Institute of Organic Chemistry and Biochemistry AS CR, v.v.i., Flemingovo nam. 2., 166 10 Prague 6, Czech Republic
| | - Brianna M. Hurysz
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92037, USA
| | - Jehad Almaliti
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, 92037, USA
| | - Yukiko Miyamoto
- Division of Gastroenterology, School of Medicine, University of California San Diego, La Jolla, CA, 92037, USA
| | - Lars Eckmann
- Division of Gastroenterology, School of Medicine, University of California San Diego, La Jolla, CA, 92037, USA
| | - William H. Gerwick
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, 92037, USA
| | - Anthony J. O’Donoghue
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92037, USA
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Evzen Boura
- Institute of Organic Chemistry and Biochemistry AS CR, v.v.i., Flemingovo nam. 2., 166 10 Prague 6, Czech Republic
| |
Collapse
|
17
|
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: 0.5] [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
|
18
|
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: 4.0] [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
|
19
|
Rosenthal PJ. Proteasome inhibitors: The next generation of antimalarial drugs? Cell Chem Biol 2023; 30:413-415. [PMID: 37207628 DOI: 10.1016/j.chembiol.2023.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/21/2023]
Abstract
Among promising targets for new antimalarials is the Plasmodium falciparum proteasome. Multiple inhibitors have demonstrated potent antimalarial activity and synergy with artemisinins. Potent irreversible peptide vinyl sulfones offer synergy, minimal resistance selection, and lack of cross-resistance. These and other proteasome inhibitors have promise as components of new combination antimalarial regimens.
Collapse
Affiliation(s)
- Philip J Rosenthal
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
20
|
Fajtova P, Hurysz BM, Miyamoto Y, Serafim M, Jiang Z, Trujillo DF, Liu L, Somani U, Almaliti J, Myers SA, Caffrey CR, Gerwick WH, Kirk CJ, Boura E, Eckmann L, O'Donoghue AJ. Development of subunit selective substrates for Trichomonas vaginalis proteasome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.05.535794. [PMID: 37066163 PMCID: PMC10104049 DOI: 10.1101/2023.04.05.535794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
The protozoan parasite, Trichomonas vaginalis (Tv) causes trichomoniasis, the most common, non-viral, sexually transmitted infection in the world. Only two closely related drugs are approved for its treatment. The accelerating emergence of resistance to these drugs and lack of alternative treatment options poses an increasing threat to public health. There is an urgent need for novel effective anti-parasitic compounds. The proteasome is a critical enzyme for T. vaginalis survival and was validated as a drug target to treat trichomoniasis. However, to develop potent inhibitors of the T. vaginalis proteasome, it is essential that we understand which subunits should be targeted. Previously, we identified two fluorogenic substrates that were cleaved by T. vaginalis proteasome, however after isolating the enzyme complex and performing an in-depth substrate specificity study, we have now designed three fluorogenic reporter substrates that are each specific for one catalytic subunit. We screened a library of peptide epoxyketone inhibitors against the live parasite and evaluated which subunits are targeted by the top hits. Together we show that targeting of the β5 subunit of T. vaginalis is sufficient to kill the parasite, however, targeting of β5 plus either β1 or β2 results in improved potency.
Collapse
|
21
|
He L, Zhang HR, Di WD, Li FF, Wang CQ, Yang X, Liu XF, Hu M. A proteasomal β5 subunit of Haemonchus contortus with a role in the growth, development and life span. Parasit Vectors 2023; 16:100. [PMID: 36922877 PMCID: PMC10015785 DOI: 10.1186/s13071-023-05676-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/18/2023] [Indexed: 03/17/2023] Open
Abstract
BACKGROUND The proteasome in eukaryotic cells can degrade a variety of proteins and plays an important role in regulating the cell cycle, cell survival and apoptosis. The proteasome receives much attention as a potential chemotherapeutic target for treatment of a variety of infectious parasitic diseases, but few studies of proteasomes have been done on parasitic nematodes. METHODS A proteasomal β5 subunit encoding gene (named Hc-pbs-5) and its inferred product (Hc-PBS-5) in Haemonchus contortus were identified and characterized in this study. Then, the transcriptional profiles and anatomical expression were studied using an integrated molecular approach. Finally, a specific proteasome inhibitor bortezomib (BTZ), together with RNA interference (RNAi), was employed to assess the function of Hc-PBS-5. RESULTS Bioinformatic analysis revealed that the coding sequence of Hc-pbs-5 was 855 bp long and encoded 284 amino acids (aa). The predicted protein (Hc-PBS-5) had core conservative sequences (65-250 aa) belonging to N-terminal nucleophile (Ntn) family of hydrolases. Real-time PCR results revealed that Hc-pbs-5 was continuously transcribed in eight developmental stages with higher levels at the infective third-stage larvae (L3s) and adult males of H. contortus. Immunohistochemical results revealed that Hc-PBS-5 was expressed in intestine, outer cuticle, muscle cells under the outer cuticle, cervical glands and seminal vesicles of male adults and also in intestine, outer cuticle, cervical glands, uterine wall, eggs and ovaries of female adults of H. contortus. BTZ could reduce proportions of egg hatching, and the fourth-stage larvae (L4s) developed from the exsheathed L3s (xL3s) of H. contortus. In addition, silencing Hc-pbs-5 by soaking the specific double-stranded RNA (dsRNA) could decrease the transcription of Hc-pbs-5 and result in fewer xL3s developing to L4s in vitro. CONCLUSIONS These results indicate that proteasomal β5 subunit plays an important role in the growth, development and life span of H. contortus.
Collapse
Affiliation(s)
- Li He
- State Key Laboratory of Agricultural Microbiology, Key Laboratory for the Development of Veterinary Products, Ministry of Agriculture College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China.,Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei Province, People's Republic of China
| | - Hong-Run Zhang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory for the Development of Veterinary Products, Ministry of Agriculture College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Wen-Da Di
- College of Animal Science and Technology, Guangxi University, Nanning, 530004, Guangxi Zhuang Autonomous Region, People's Republic of China.
| | - Fang-Fang Li
- State Key Laboratory of Agricultural Microbiology, Key Laboratory for the Development of Veterinary Products, Ministry of Agriculture College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Chun-Qun Wang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory for the Development of Veterinary Products, Ministry of Agriculture College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Xin Yang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory for the Development of Veterinary Products, Ministry of Agriculture College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Xiao-Fang Liu
- State Key Laboratory of Agricultural Microbiology, Key Laboratory for the Development of Veterinary Products, Ministry of Agriculture College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China
| | - Min Hu
- State Key Laboratory of Agricultural Microbiology, Key Laboratory for the Development of Veterinary Products, Ministry of Agriculture College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei Province, People's Republic of China.
| |
Collapse
|
22
|
Espinoza-Chávez R, Salerno A, Liuzzi A, Ilari A, Milelli A, Uliassi E, Bolognesi ML. Targeted Protein Degradation for Infectious Diseases: from Basic Biology to Drug Discovery. ACS BIO & MED CHEM AU 2023; 3:32-45. [PMID: 37101607 PMCID: PMC10125329 DOI: 10.1021/acsbiomedchemau.2c00063] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/04/2022] [Accepted: 11/23/2022] [Indexed: 04/28/2023]
Abstract
Targeted protein degradation (TPD) is emerging as one of the most innovative strategies to tackle infectious diseases. Particularly, proteolysis-targeting chimera (PROTAC)-mediated protein degradation may offer several benefits over classical anti-infective small-molecule drugs. Because of their peculiar and catalytic mechanism of action, anti-infective PROTACs might be advantageous in terms of efficacy, toxicity, and selectivity. Importantly, PROTACs may also overcome the emergence of antimicrobial resistance. Furthermore, anti-infective PROTACs might have the potential to (i) modulate "undruggable" targets, (ii) "recycle" inhibitors from classical drug discovery approaches, and (iii) open new scenarios for combination therapies. Here, we try to address these points by discussing selected case studies of antiviral PROTACs and the first-in-class antibacterial PROTACs. Finally, we discuss how the field of PROTAC-mediated TPD might be exploited in parasitic diseases. Since no antiparasitic PROTAC has been reported yet, we also describe the parasite proteasome system. While in its infancy and with many challenges ahead, we hope that PROTAC-mediated protein degradation for infectious diseases may lead to the development of next-generation anti-infective drugs.
Collapse
Affiliation(s)
- Rocío
Marisol Espinoza-Chávez
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Alessandra Salerno
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Anastasia Liuzzi
- Institute
of Molecular Biology and Pathology of the Italian National Research
Council (IBPM-CNR) - Department of Biochemical Sciences, Sapienza University, P.le A. Moro 5, 00185 Roma, Italy
| | - Andrea Ilari
- Institute
of Molecular Biology and Pathology of the Italian National Research
Council (IBPM-CNR) - Department of Biochemical Sciences, Sapienza University, P.le A. Moro 5, 00185 Roma, Italy
| | - Andrea Milelli
- Department
for Life Quality Studies, Alma Mater Studiorum
- University of Bologna, Corso d’Augusto 237, 47921 Rimini, Italy
| | - Elisa Uliassi
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Maria Laura Bolognesi
- Department
of Pharmacy and Biotechnology, Alma Mater
Studiorum - University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| |
Collapse
|
23
|
Koester DC, Marx VM, Williams S, Jiricek J, Dauphinais M, René O, Miller SL, Zhang L, Patra D, Chen YL, Cheung H, Gable J, Lakshminarayana SB, Osborne C, Galarneau JR, Kulkarni U, Richmond W, Bretz A, Xiao L, Supek F, Wiesmann C, Honnappa S, Be C, Mäser P, Kaiser M, Ritchie R, Barrett MP, Diagana TT, Sarko C, Rao SPS. Discovery of Novel Quinoline-Based Proteasome Inhibitors for Human African Trypanosomiasis (HAT). J Med Chem 2022; 65:11776-11787. [PMID: 35993839 PMCID: PMC9469205 DOI: 10.1021/acs.jmedchem.2c00791] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Human African Trypanosomiasis (HAT) is a vector-borne disease caused by kinetoplastid parasites of the Trypanosoma genus. The disease proceeds in two stages, with a hemolymphatic blood stage and a meningo-encephalic brain stage. In the latter stage, the parasite causes irreversible damage to the brain leading to sleep cycle disruption and is fatal if untreated. An orally bioavailable treatment is highly desirable. In this study, we present a brain-penetrant, parasite-selective 20S proteasome inhibitor that was rapidly optimized from an HTS singleton hit to drug candidate compound 7 that showed cure in a stage II mouse efficacy model. Here, we describe hit expansion and lead optimization campaign guided by cryo-electron microscopy and an in silico model to predict the brain-to-plasma partition coefficient Kp as an important parameter to prioritize compounds for synthesis. The model combined with in vitro and in vivo experiments allowed us to advance compounds with favorable unbound brain-to-plasma ratios (Kp,uu) to cure a CNS disease such as HAT.
Collapse
Affiliation(s)
- Dennis C. Koester
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Vanessa M. Marx
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Sarah Williams
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Jan Jiricek
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Maxime Dauphinais
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Olivier René
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Sarah L. Miller
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Lei Zhang
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Debjani Patra
- Novartis
Institutes for Tropical Diseases, Emeryville, California 94608, United States
| | - Yen-Liang Chen
- Lead
Discovery, Novartis Institutes for Tropical
Diseases, Emeryville, California 94608, United States
| | - Harry Cheung
- Lead
Discovery, Novartis Institutes for Tropical
Diseases, Emeryville, California 94608, United States
| | - Jonathan Gable
- Lead
Discovery, Novartis Institutes for Tropical
Diseases, Emeryville, California 94608, United States
| | - Suresh B. Lakshminarayana
- Pharmacokinetic
Sciences, Novartis Institutes for Tropical
Diseases, Emeryville, California 94608, United States
| | - Colin Osborne
- Pharmacokinetic
Sciences, Pharmacology and Comparative Medicine, Novartis Institutes for Tropical Diseases, Emeryville, California 94608, United States
| | - Jean-Rene Galarneau
- Preclinical
Safety, Novartis Institutes for Biomedical
Research, Cambridge, Massachusetts 02139, United States
| | - Upendra Kulkarni
- Chemical
and Pharmaceutical Profiling, Novartis Institutes
for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Wendy Richmond
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, San Diego, California 92121, United States
| | - Angela Bretz
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, San Diego, California 92121, United States
| | - Linda Xiao
- Pharmacology, Novartis Institutes for Tropical Diseases, Emeryville, California 94608, United States
| | - Frantisek Supek
- Novartis
Institutes for Biomedical Research, San Diego, California 92121, United States
| | | | - Srinivas Honnappa
- Novartis
Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Celine Be
- Novartis
Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- University
of Basel, CH 4000 Basel, Switzerland
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- University
of Basel, CH 4000 Basel, Switzerland
| | - Ryan Ritchie
- University of Glasgow, University Place, Glasgow G12 8TA, U.K
| | | | - Thierry T. Diagana
- Novartis
Institutes for Tropical Diseases, Emeryville, California 94608, United States
| | - Christopher Sarko
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Srinivasa P. S. Rao
- Novartis
Institutes for Tropical Diseases, Emeryville, California 94608, United States
| |
Collapse
|
24
|
Wu J, Zhang F, Tao H, Nawaz W, Chen D, Wu Z. The potential roles of interleukin-25 in infectious diseases. Front Immunol 2022; 13:986118. [PMID: 36119076 PMCID: PMC9478665 DOI: 10.3389/fimmu.2022.986118] [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: 07/04/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Interleukin-25 (IL-25), also known as IL-17E, is a recently identified cytokine of the IL-17 family. Numerous studies illustrated that the expression of IL-25 is regulated by multiple pathogens, including parasitic, viral, and bacterial infections. IL-25 has a dual function in infectious diseases. On the one hand, IL-25 activates type 2 immunity via the relevant cytokines, including IL-4, IL-5, and IL-13, which are associated with the development of pathogenic infection-related allergic diseases. On the other hand, IL-25 involves in the recruitment of group 2 innate lymphoid cells (ILC2) to enhanced T helper 2 (Th2) cell differentiation, which are important to the clearance of pathogens. However, the precise roles of IL-25 in infectious diseases remain largely unknown. Thus, the current review will shed light on the pivotal roles of IL-25 in infectious diseases.
Collapse
Affiliation(s)
- Jing Wu
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Fang Zhang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Hongji Tao
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
| | - Waqas Nawaz
- Hôpital Maisonneuve-Rosemont, School of Medicine, University of Montreal, Montréal, Canada
| | - Deyan Chen
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
- *Correspondence: Deyan Chen, ; Zhiwei Wu,
| | - Zhiwei Wu
- Center for Public Health Research, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
- *Correspondence: Deyan Chen, ; Zhiwei Wu,
| |
Collapse
|
25
|
Almeida-Silva J, Menezes DS, Fernandes JMP, Almeida MC, Vasco-Dos-Santos DR, Saraiva RM, Viçosa AL, Perez SAC, Andrade SG, Suarez-Fontes AM, Vannier-Santos MA. The repositioned drugs disulfiram/diethyldithiocarbamate combined to benznidazole: Searching for Chagas disease selective therapy, preventing toxicity and drug resistance. Front Cell Infect Microbiol 2022; 12:926699. [PMID: 35967878 PMCID: PMC9372510 DOI: 10.3389/fcimb.2022.926699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/27/2022] [Indexed: 12/20/2022] Open
Abstract
Chagas disease (CD) affects at least 6 million people in 21 South American countries besides several thousand in other nations all over the world. It is estimated that at least 14,000 people die every year of CD. Since vaccines are not available, chemotherapy remains of pivotal relevance. About 30% of the treated patients cannot complete the therapy because of severe adverse reactions. Thus, the search for novel drugs is required. Here we tested the benznidazole (BZ) combination with the repositioned drug disulfiram (DSF) and its derivative diethyldithiocarbamate (DETC) upon Trypanosoma cruzi in vitro and in vivo. DETC-BZ combination was synergistic diminishing epimastigote proliferation and enhancing selective indexes up to over 10-fold. DETC was effective upon amastigotes of the BZ- partially resistant Y and the BZ-resistant Colombiana strains. The combination reduced proliferation even using low concentrations (e.g., 2.5 µM). Scanning electron microscopy revealed membrane discontinuities and cell body volume reduction. Transmission electron microscopy revealed remarkable enlargement of endoplasmic reticulum cisternae besides, dilated mitochondria with decreased electron density and disorganized kinetoplast DNA. At advanced stages, the cytoplasm vacuolation apparently impaired compartmentation. The fluorescent probe H2-DCFDA indicates the increased production of reactive oxygen species associated with enhanced lipid peroxidation in parasites incubated with DETC. The biochemical measurement indicates the downmodulation of thiol expression. DETC inhibited superoxide dismutase activity on parasites was more pronounced than in infected mice. In order to approach the DETC effects on intracellular infection, peritoneal macrophages were infected with Colombiana trypomastigotes. DETC addition diminished parasite numbers and the DETC-BZ combination was effective, despite the low concentrations used. In the murine infection, the combination significantly enhanced animal survival, decreasing parasitemia over BZ. Histopathology revealed that low doses of BZ-treated animals presented myocardial amastigote, not observed in combination-treated animals. The picrosirius collagen staining showed reduced myocardial fibrosis. Aminotransferase de aspartate, Aminotransferase de alanine, Creatine kinase, and urea plasma levels demonstrated that the combination was non-toxic. As DSF and DETC can reduce the toxicity of other drugs and resistance phenotypes, such a combination may be safe and effective.
Collapse
Affiliation(s)
- Juliana Almeida-Silva
- Innovations in Therapies, Education and Bioproducts Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Diego Silva Menezes
- Parasite Biology Laboratory, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, BA, Brazil
| | - Juan Mateus Pereira Fernandes
- Innovations in Therapies, Education and Bioproducts Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Márcio Cerqueira Almeida
- Parasite Biology Laboratory, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, BA, Brazil
| | - Deyvison Rhuan Vasco-Dos-Santos
- Innovations in Therapies, Education and Bioproducts Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Roberto Magalhães Saraiva
- Laboratory of Clinical Research on Chagas Disease, Evandro Chagas Infectious Disease Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Alessandra Lifsitch Viçosa
- Experimental Pharmacotechnics Laboratory, Department of Galenic Innovation, Institute of Drug Technology - Farmanguinhos, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Sandra Aurora Chavez Perez
- Project Management Technical Assistance, Institute of Drug Technology - Farmanguinhos, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Sônia Gumes Andrade
- Experimental Chagas Disease Laboratory, Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, BA, Brazil
| | - Ana Márcia Suarez-Fontes
- Innovations in Therapies, Education and Bioproducts Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Marcos André Vannier-Santos
- Innovations in Therapies, Education and Bioproducts Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
26
|
Jin B, Li G, Zhou L, Fan Z. Mechanism Involved in Acute Liver Injury Induced by Intestinal Ischemia-Reperfusion. Front Pharmacol 2022; 13:924695. [PMID: 35694264 PMCID: PMC9185410 DOI: 10.3389/fphar.2022.924695] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/05/2022] [Indexed: 12/28/2022] Open
Abstract
Intestinal ischemia-reperfusion (I/R) is a common pathophysiological process, which can occur in many conditions such as acute enteric ischemia, severe burns, small intestinal transplantation, etc,. Ischemia-reperfusion of the intestine is often accompanied by distal organ injury, especially liver injury. This paper outlined the signal pathways and cytokines involved in acute liver injury induced by intestinal I/R: the NF-κB Signaling Pathway, the P66shc Signaling Pathway, the HMGB1 Signaling Pathway, the Nrf2-ARE Signaling Pathway, the AMPK-SIRT-1 Signaling Pathway and other cytokines, providing new ideas for the prevention and treatment of liver injury caused by reperfusion after intestinal I/R.
Collapse
Affiliation(s)
- Binghui Jin
- Department of General Surgery, The Third People’s Hospital of Dalian, Dalian Medical University, Dalian, China
- Department of Central Laboratory, The Third People’s Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Guangyao Li
- Department of General Surgery, The Third People’s Hospital of Dalian, Dalian Medical University, Dalian, China
- Department of Central Laboratory, The Third People’s Hospital of Dalian, Dalian Medical University, Dalian, China
| | - Lin Zhou
- Department of Outpatient, the NO. 967 Hospital of PLA Joint Logistics Support Force, Dalian Medical University, Dalian, China
| | - Zhe Fan
- Department of General Surgery, The Third People’s Hospital of Dalian, Dalian Medical University, Dalian, China
- Department of Central Laboratory, The Third People’s Hospital of Dalian, Dalian Medical University, Dalian, China
| |
Collapse
|
27
|
Kobakhidze G, Sethi A, Valimehr S, Ralph SA, Rouiller I. The AAA+ ATPase p97 as a novel parasite and tuberculosis drug target. Trends Parasitol 2022; 38:572-590. [DOI: 10.1016/j.pt.2022.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 10/18/2022]
|
28
|
Imran M, Khan SA, Abida, Alshrari AS, Eltahir Mudawi MM, Alshammari MK, Harshan AA, Alshammari NA. Small molecules as kinetoplastid specific proteasome inhibitors for Leishmaniasis: a patent review from 1998 to 2021. Expert Opin Ther Pat 2022; 32:591-604. [PMID: 35220857 DOI: 10.1080/13543776.2022.2045948] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION : Leishmaniasis is a neglected tropical infectious disease. The available limited therapeutic options for leishmaniasis are inadequate due to their poor pharmacokinetic profile, resistance, toxicity, high cost, and compliance problems. This warrants identification of new targets for the development of safer and effective anti-Leishmania therapy. The kinetoplastid specific proteasome (KSP) is a novel validated target to develop drugs against leishmaniasis. AREA COVERED : This review focuses on all the published patent applications and granted patents related to the studied small molecules as KSP inhibitors (KSPIs) against Leishmania from 1998 to December 31, 2021. EXPERT OPINION : A little amount of work has been done on KSPIs, but the study results are quite encouraging. LXE408 and GSK3494245 are two KSPIs in different phases of clinical trials. Some other small molecules have also shown KSP inhibitory potential, but they are not in clinical trials. The KSPIs are promising next-generation orally active patient compliant drugs against kinetoplastid diseases, including leishmaniasis. However, the main challenge to discover the KSPIs will be the resistance development and their selectivity against the proteasome of eukaryotic cells.
Collapse
Affiliation(s)
- Mohd Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Shah Alam Khan
- College of Pharmacy, National University of Science and Technology, Muscat 130, Oman
| | - Abida
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Ahmed Subeh Alshrari
- Medical Laboratory Technology Department, Faculty of Applied Medical Science, Northern Border University, Arar 91431, Saudi Arabia
| | | | - Mohammed Kanan Alshammari
- Department of Pharmaceutical Care, Rafha Central Hospital, North Zone, Rafha 91911, Kingdom of Saudi Arabia
| | - Aishah Ali Harshan
- Department of Pharmaceutical Care, Northern Area Armed Forces Hospital, King Khalid Military City Hospital, Hafr Al-Batin, Kingdom of Saudi Arabia
| | - Noufah Aqeel Alshammari
- Department of Pharmaceutical Care, Security Forces Hospital, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
29
|
Kourbeli V, Chontzopoulou E, Moschovou K, Pavlos D, Mavromoustakos T, Papanastasiou IP. An Overview on Target-Based Drug Design against Kinetoplastid Protozoan Infections: Human African Trypanosomiasis, Chagas Disease and Leishmaniases. Molecules 2021; 26:molecules26154629. [PMID: 34361781 PMCID: PMC8348971 DOI: 10.3390/molecules26154629] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 02/06/2023] Open
Abstract
The protozoan diseases Human African Trypanosomiasis (HAT), Chagas disease (CD), and leishmaniases span worldwide and therefore their impact is a universal concern. The present regimen against kinetoplastid protozoan infections is poor and insufficient. Target-based design expands the horizon of drug design and development and offers novel chemical entities and potential drug candidates to the therapeutic arsenal against the aforementioned neglected diseases. In this review, we report the most promising targets of the main kinetoplastid parasites, as well as their corresponding inhibitors. This overview is part of the Special Issue, entitled "Advances of Medicinal Chemistry against Kinetoplastid Protozoa (Trypanosoma brucei, Trypanosoma cruzi and Leishmania spp.) Infections: Drug Design, Synthesis and Pharmacology".
Collapse
Affiliation(s)
- Violeta Kourbeli
- Department of Pharmacy, Division of Pharmaceutical Chemistry, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupoli-Zografou, 157 84 Athens, Greece;
| | - Eleni Chontzopoulou
- Department of Organic Chemistry, Faculty of Chemistry, National and Kapodistrian University of Athens, Panepistimioupoli-Zografou, 157 71 Athens, Greece; (E.C.); (K.M.); (D.P.); (T.M.)
| | - Kalliopi Moschovou
- Department of Organic Chemistry, Faculty of Chemistry, National and Kapodistrian University of Athens, Panepistimioupoli-Zografou, 157 71 Athens, Greece; (E.C.); (K.M.); (D.P.); (T.M.)
| | - Dimitrios Pavlos
- Department of Organic Chemistry, Faculty of Chemistry, National and Kapodistrian University of Athens, Panepistimioupoli-Zografou, 157 71 Athens, Greece; (E.C.); (K.M.); (D.P.); (T.M.)
| | - Thomas Mavromoustakos
- Department of Organic Chemistry, Faculty of Chemistry, National and Kapodistrian University of Athens, Panepistimioupoli-Zografou, 157 71 Athens, Greece; (E.C.); (K.M.); (D.P.); (T.M.)
| | - Ioannis P. Papanastasiou
- Department of Pharmacy, Division of Pharmaceutical Chemistry, School of Health Sciences, National and Kapodistrian University of Athens, Panepistimioupoli-Zografou, 157 84 Athens, Greece;
- Correspondence:
| |
Collapse
|
30
|
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: 1.0] [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
|
31
|
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
|
32
|
Shrivastav MT, Malik Z, Somlata. Revisiting Drug Development Against the Neglected Tropical Disease, Amebiasis. Front Cell Infect Microbiol 2021; 10:628257. [PMID: 33718258 PMCID: PMC7943716 DOI: 10.3389/fcimb.2020.628257] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/30/2020] [Indexed: 11/15/2022] Open
Abstract
Amebiasis is a neglected tropical disease which is caused by the protozoan parasite Entamoeba histolytica. This disease is one of the leading causes of diarrhea globally, affecting largely impoverished residents in developing countries. Amebiasis also remains one of the top causes of gastrointestinal diseases in returning international travellers. Despite having many side effects, metronidazole remains the drug of choice as an amebicidal tissue-active agent. However, emergence of metronidazole resistance in pathogens having similar anaerobic metabolism and also in laboratory strains of E. histolytica has necessitated the identification and development of new drug targets and therapeutic strategies against the parasite. Recent research in the field of amebiasis has led to a better understanding of the parasite’s metabolic and cellular pathways and hence has been useful in identifying new drug targets. On the other hand, new molecules effective against amebiasis have been mined by modifying available compounds, thereby increasing their potency and efficacy and also by repurposing existing approved drugs. This review aims at compiling and examining up to date information on promising drug targets and drug molecules for the treatment of amebiasis.
Collapse
Affiliation(s)
- Manish T Shrivastav
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Zainab Malik
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Somlata
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
33
|
20S proteasomes secreted by the malaria parasite promote its growth. Nat Commun 2021; 12:1172. [PMID: 33608523 PMCID: PMC7895969 DOI: 10.1038/s41467-021-21344-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/20/2021] [Indexed: 12/16/2022] Open
Abstract
Mature red blood cells (RBCs) lack internal organelles and canonical defense mechanisms, making them both a fascinating host cell, in general, and an intriguing choice for the deadly malaria parasite Plasmodium falciparum (Pf), in particular. Pf, while growing inside its natural host, the human RBC, secretes multipurpose extracellular vesicles (EVs), yet their influence on this essential host cell remains unknown. Here we demonstrate that Pf parasites, cultured in fresh human donor blood, secrete within such EVs assembled and functional 20S proteasome complexes (EV-20S). The EV-20S proteasomes modulate the mechanical properties of naïve human RBCs by remodeling their cytoskeletal network. Furthermore, we identify four degradation targets of the secreted 20S proteasome, the phosphorylated cytoskeletal proteins β-adducin, ankyrin-1, dematin and Epb4.1. Overall, our findings reveal a previously unknown 20S proteasome secretion mechanism employed by the human malaria parasite, which primes RBCs for parasite invasion by altering membrane stiffness, to facilitate malaria parasite growth. Plasmodium falciparum secretes extracellular vesicles (EVs) while growing inside red blood cells (RBCs). Here the authors show that these EVs contain assembled and functional 20S proteasome complexes that remodel the cytoskeleton of naïve human RBCs, priming the RBCs for parasite invasion.
Collapse
|
34
|
Thompson AM, O’Connor PD, Yardley V, Maes L, Launay D, Braillard S, Chatelain E, Wan B, Franzblau SG, Ma Z, Cooper CB, Denny WA. Novel Linker Variants of Antileishmanial/Antitubercular 7-Substituted 2-Nitroimidazooxazines Offer Enhanced Solubility. ACS Med Chem Lett 2021; 12:275-281. [PMID: 33603975 DOI: 10.1021/acsmedchemlett.0c00649] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 01/15/2021] [Indexed: 12/15/2022] Open
Abstract
Antitubercular 7-substituted 2-nitroimidazo[2,1-b][1,3]oxazines were previously shown to exhibit potent antileishmanial and antitrypanosomal activities, culminating in a new clinical investigational drug for visceral leishmaniasis (DNDI-0690). To offset development risks, we continued to seek further leads with divergent candidate profiles, especially analogues possessing greater aqueous solubility. Starting from an efficacious monoaryl derivative, replacement of the side chain ether linkage by novel amine, amide, and urea functionality was first explored; the former substitution was well-tolerated in vitro and in vivo but elicited marginal alterations to solubility (except through a less stable benzylamine), whereas the latter groups resulted in significant solubility improvements (up to 53-fold) but an antileishmanial potency reduction of at least 10-fold. Ultimately, we discovered that O-carbamate 66 offered a more optimal balance of increased solubility, suitable metabolic stability, excellent oral bioavailability (100%), and strong in vivo efficacy in a visceral leishmaniasis mouse model (97% parasite load reduction at 25 mg/kg).
Collapse
Affiliation(s)
- Andrew M. Thompson
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Patrick D. O’Connor
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Vanessa Yardley
- Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - Louis Maes
- Laboratory for Microbiology, Parasitology and Hygiene, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Delphine Launay
- Drugs for Neglected Diseases Initiative, 15 Chemin Louis Dunant, 1202 Geneva, Switzerland
| | - Stephanie Braillard
- Drugs for Neglected Diseases Initiative, 15 Chemin Louis Dunant, 1202 Geneva, Switzerland
| | - Eric Chatelain
- Drugs for Neglected Diseases Initiative, 15 Chemin Louis Dunant, 1202 Geneva, Switzerland
| | - Baojie Wan
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612, United States
| | - Scott G. Franzblau
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 South Wood Street, Chicago, Illinois 60612, United States
| | - Zhenkun Ma
- Global Alliance for TB Drug Development, 40 Wall Street, New York, New York 10005, United States
| | - Christopher B. Cooper
- Global Alliance for TB Drug Development, 40 Wall Street, New York, New York 10005, United States
| | - William A. Denny
- Auckland Cancer Society Research Centre, School of Medical Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
35
|
Tukulula M, Louw S, Njoroge M, Chibale K. Synthesis and In Vitro Antiprotozoan Evaluation of 4-/8-Aminoquinoline-based Lactams and Tetrazoles. Molecules 2020; 25:E5941. [PMID: 33333924 PMCID: PMC7765388 DOI: 10.3390/molecules25245941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/18/2020] [Accepted: 11/25/2020] [Indexed: 11/16/2022] Open
Abstract
A second generation of 4-aminoquinoline- and 8-aminoquinoline-based tetrazoles and lactams were synthesized via the Staudinger and Ugi multicomponent reactions. These compounds were subsequently evaluated in vitro for their potential antiplasmodium activity against a multidrug-resistant K1 strain and for their antitrypanosomal activity against a cultured T. b. rhodesiense STIB900 strain. Several of these compounds (4a-g) displayed good antiplasmodium activities (IC50 = 0.20-0.62 µM) that were comparable to the reference drugs, while their antitrypanosomal activity was moderate (<20 µM). Compound 4e was 2-fold more active than primaquine and was also the most active (IC50 = 7.01 µM) against T. b. rhodesiense and also exhibited excellent aqueous solubility (>200 µM) at pH 7.
Collapse
Affiliation(s)
| | - Stefan Louw
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa; (S.L.); (M.N.); (K.C.)
- Department of Chemistry and Biochemistry, University of Namibia, Windhoek 10005, Namibia
| | - Mathew Njoroge
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa; (S.L.); (M.N.); (K.C.)
| | - Kelly Chibale
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa; (S.L.); (M.N.); (K.C.)
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|
36
|
Hubbell GE, Tepe JJ. Natural product scaffolds as inspiration for the design and synthesis of 20S human proteasome inhibitors. RSC Chem Biol 2020; 1:305-332. [PMID: 33791679 PMCID: PMC8009326 DOI: 10.1039/d0cb00111b] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022] Open
Abstract
The 20S proteasome is a valuable target for the treatment of a number of diseases including cancer, neurodegenerative disease, and parasitic infection. In an effort to discover novel inhibitors of the 20S proteasome, many reseaarchers have looked to natural products as potential leads for drug discovery. The following review discusses the efforts made in the field to isolate and identify natural products as inhibitors of the proteasome. In addition, we describe some of the modifications made to natural products in order to discover more potent and selective inhibitors for potential disease treatment.
Collapse
Affiliation(s)
- Grace E. Hubbell
- Department of Chemistry, Michigan State UniversityEast LansingMI 48823USA
| | - Jetze J. Tepe
- Department of Chemistry, Michigan State UniversityEast LansingMI 48823USA
| |
Collapse
|
37
|
Cao Y, Zhu H, He R, Kong L, Shao J, Zhuang R, Xi J, Zhang J. Proteasome, a Promising Therapeutic Target for Multiple Diseases Beyond Cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:4327-4342. [PMID: 33116419 PMCID: PMC7585272 DOI: 10.2147/dddt.s265793] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/22/2020] [Indexed: 12/14/2022]
Abstract
Proteasome is vital for intracellular protein homeostasis as it eliminates misfolded and damaged protein. Inhibition of proteasome has been validated as a powerful strategy for anti-cancer therapy, and several drugs have been approved for treatment of multiple myeloma. Recent studies indicate that proteasome has potent therapeutic effects on a variety of diseases besides cancer, including parasite infectious diseases, bacterial/fungal infections diseases, neurodegenerative diseases and autoimmune diseases. In this review, recent developments of proteasome inhibitors for various diseases and related structure activity relationships are going to be summarized.
Collapse
Affiliation(s)
- Yu Cao
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang Province, 310015, People's Republic of China
| | - Huajian Zhu
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang Province, 310015, People's Republic of China
| | - Ruoyu He
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, Zhejiang Province, 310023 People's Republic of China
| | - Limin Kong
- Department of Pharmacy, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang Province, 310003, People's Republic of China
| | - Jiaan Shao
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang Province, 310015, People's Republic of China
| | - Rangxiao Zhuang
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, Zhejiang Province, 310023 People's Republic of China
| | - Jianjun Xi
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, Zhejiang Province, 310023 People's Republic of China
| | - Jiankang Zhang
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang Province, 310015, People's Republic of China
| |
Collapse
|
38
|
Ghosh S, Farr L, Singh A, Leaton LA, Padalia J, Shirley DA, Sullivan D, Moonah S. COP9 signalosome is an essential and druggable parasite target that regulates protein degradation. PLoS Pathog 2020; 16:e1008952. [PMID: 32960936 PMCID: PMC7531848 DOI: 10.1371/journal.ppat.1008952] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 10/02/2020] [Accepted: 09/01/2020] [Indexed: 02/06/2023] Open
Abstract
Understanding how the protozoan protein degradation pathway is regulated could uncover new parasite biology for drug discovery. We found the COP9 signalosome (CSN) conserved in multiple pathogens such as Leishmania, Trypanosoma, Toxoplasma, and used the severe diarrhea-causing Entamoeba histolytica to study its function in medically significant protozoa. We show that CSN is an essential upstream regulator of parasite protein degradation. Genetic disruption of E. histolytica CSN by two distinct approaches inhibited cell proliferation and viability. Both CSN5 knockdown and dominant negative mutation trapped cullin in a neddylated state, disrupting UPS activity and protein degradation. In addition, zinc ditiocarb (ZnDTC), a main metabolite of the inexpensive FDA-approved globally-available drug disulfiram, was active against parasites acting in a COP9-dependent manner. ZnDTC, given as disulfiram-zinc, had oral efficacy in clearing parasites in vivo. Our findings provide insights into the regulation of parasite protein degradation, and supports the significant therapeutic potential of COP9 inhibition.
Collapse
Affiliation(s)
- Swagata Ghosh
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Laura Farr
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Aditya Singh
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Laura-Ann Leaton
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado, Aurora, CO, United States of America
| | - Jay Padalia
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Debbie-Ann Shirley
- Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - David Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States of America
| | - Shannon Moonah
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| |
Collapse
|