1
|
Silva GM, Gomes SQ, Lopes CD, de Albuquerque S, de Paula da Silva CHT. Structural analysis and shape-based identification of novel inhibitors targeting the Trypanosoma cruzi proteasome. Int J Biol Macromol 2024; 277:134290. [PMID: 39084432 DOI: 10.1016/j.ijbiomac.2024.134290] [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: 06/13/2024] [Revised: 07/17/2024] [Accepted: 07/28/2024] [Indexed: 08/02/2024]
Abstract
There is an urgent need to develop new, safer, and more effective drugs against Chagas disease (CD) as well as related kinetoplastid diseases. Targeting and inhibiting the Trypanosoma cruzi proteasome has emerged as a promising therapeutic approach in this context. To expand the chemical space for this class of inhibitors, we performed virtual screening campaigns with emphasis on shape-based similarity and ADMET prioritization. We describe the ideation and application of robustly validated shape queries for these campaigns, which furnished 44 compounds for biological evaluation. Five hit compounds demonstrated in vitro antitrypanosomal activity by potential inhibition of T. cruzi proteasome and notable chemical diversities, particularly, LCQFTC11. Structural insights were achieved by homology modeling, sequence/structure alignment, proteasome-species comparison, docking, molecular dynamics, and MMGBSA binding affinity estimations. These methods confirmed key interactions as well as the stability of LCQFTC11 at the β4/β5 subunits' binding site of the T. cruzi proteasome, consistent with known inhibitors. Our results warrant future assay confirmation of our hit as a T. cruzi proteasome inhibitor. Importantly, we also shed light into dynamic details for a proteasome inhibition mechanism that shall be further investigated. We expect to contribute to the development of viable CD drug candidates through such a relevant approach.
Collapse
Affiliation(s)
- Guilherme Martins Silva
- Center for Drug Discovery and Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA; Computational Laboratory of Pharmaceutical Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, Ribeirão Preto, SP 14040-903, Brazil.
| | - Suzane Quintana Gomes
- Computational Laboratory of Pharmaceutical Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, Ribeirão Preto, SP 14040-903, Brazil
| | - Carla Duque Lopes
- Laboratório de Parasitologia, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, Ribeirão Preto, SP 14040-903, Brazil
| | - Sérgio de Albuquerque
- Laboratório de Parasitologia, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, Ribeirão Preto, SP 14040-903, Brazil
| | - Carlos Henrique Tomich de Paula da Silva
- Computational Laboratory of Pharmaceutical Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Av. do Café, s/n, Ribeirão Preto, SP 14040-903, Brazil
| |
Collapse
|
2
|
Safaei M, Goodarzi A, Abpeikar Z, Farmani AR, Kouhpayeh SA, Najafipour S, Jafari Najaf Abadi MH. Determination of key hub genes in Leishmaniasis as potential factors in diagnosis and treatment based on a bioinformatics study. Sci Rep 2024; 14:22537. [PMID: 39342024 PMCID: PMC11438978 DOI: 10.1038/s41598-024-73779-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: 01/23/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024] Open
Abstract
Leishmaniasis is an infectious disease caused by protozoan parasites from different species of leishmania. The disease is transmitted by female sandflies that carry these parasites. In this study, datasets on leishmaniasis published in the GEO database were analyzed and summarized. The analysis in all three datasets (GSE43880, GSE55664, and GSE63931) used in this study has been performed on the skin wounds of patients infected with a clinical form of leishmania (Leishmania braziliensis), and biopsies have been taken from them. To identify differentially expressed genes (DEGs) between leishmaniasis patients and controls, the robust rank aggregation (RRA) procedure was applied. We performed gene functional annotation and protein-protein interaction (PPI) network analysis to demonstrate the putative functionalities of the DEGs. The study utilized Molecular Complex Detection (MCODE), Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) to detect molecular complexes within the protein-protein interaction (PPI) network and conduct analyses on the identified functional modules. The CytoHubba plugin's results were paired with RRA analysis to determine the hub genes. Finally, the interaction between miRNAs and hub genes was predicted. Based on the RRA integrated analysis, 407 DEGs were identified (263 up-regulated genes and 144 down-regulated genes). The top three modules were listed after creating the PPI network via the MCODE plug. Seven hub genes were found using the CytoHubba app and RRA: CXCL10, GBP1, GNLY, GZMA, GZMB, NKG7, and UBD. According to our enrichment analysis, these functional modules were primarily associated with immune pathways, cytokine activity/signaling pathways, and inflammation pathways. However, a UBD hub gene is interestingly involved in the ubiquitination pathways of pathogenesis. The mirNet database predicted the hub gene's interaction with miRNAs, and results revealed that several miRNAs, including mir-146a-5p, crucial in fighting pathogenesis. The key hub genes discovered in this work may be considered as potential biomarkers in diagnosis, development of agonists/antagonist, novel vaccine design, and will greatly contribute to clinical studies in the future.
Collapse
Affiliation(s)
- Mohsen Safaei
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Arash Goodarzi
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Zahra Abpeikar
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran.
| | - Ahmad Reza Farmani
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Seyed Amin Kouhpayeh
- Department of Pharmacology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Sohrab Najafipour
- Department of Microbiology, Faculty of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Mohammad Hassan Jafari Najaf Abadi
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran.
- Research Center for Health Technology Assessment and Medical Informatics, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
3
|
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
|
4
|
Queffeulou M, Leprohon P, Fernandez-Prada C, Ouellette M, Mejía-Jaramillo AM. CRISPR-Cas9 high-throughput screening to study drug resistance in Leishmania infantum. mBio 2024; 15:e0047724. [PMID: 38864609 PMCID: PMC11253630 DOI: 10.1128/mbio.00477-24] [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: 02/14/2024] [Accepted: 04/23/2024] [Indexed: 06/13/2024] Open
Abstract
Parasites of the genus Leishmania pose a global health threat with limited treatment options. New drugs are urgently needed, and genomic screens have the potential to accelerate target discovery, mode of action, and resistance mechanisms against these new drugs. We describe here our effort in developing a genome-wide CRISPR-Cas9 screen in Leishmania, an organism lacking a functional nonhomologous end joining system that must rely on microhomology-mediated end joining, single-strand annealing, or homologous recombination for repairing Cas9-induced double-stranded DNA breaks. A new vector for cloning and expressing single guide RNAs (sgRNAs) was designed and proven to be effective in a small pilot project while enriching specific sgRNAs during drug selection. We then developed a whole-genome library of 49,754 sgRNAs, targeting all the genes of Leishmania infantum. This library was transfected in L. infantum expressing Cas9, and these cells were selected for resistance to two antileishmanials, miltefosine and amphotericin B. The sgRNAs the most enriched in the miltefosine screen targeted the miltefosine transporter gene, but sgRNAs targeting genes coding for a RING-variant protein and a transmembrane protein were also enriched. The sgRNAs the most enriched by amphotericin B targeted the sterol 24 C methyltransferase genes and a hypothetical gene. Through gene disruption experiments, we proved that loss of function of these genes was associated with resistance. This study describes the feasibility of carrying out whole-genome CRISPR-Cas9 screens in Leishmania provided that a strong selective pressure is applied. Such a screen can be used for accelerating the development of urgently needed antileishmanial drugs.IMPORTANCELeishmaniasis, a global health threat, lacks adequate treatment options and drug resistance exacerbates the challenge. This study introduces a CRISPR-Cas9 screening approach in Leishmania infantum, unraveling mechanisms of drug resistance at a genome-wide scale. Our screen was applied against two main antileishmanial drugs, and guides were enriched upon drug selection. These guides targeted known and new targets, hence validating the use of this screen against Leishmania. This strategy provides a powerful tool to expedite drug discovery as well as potential therapeutic targets against this neglected tropical disease.
Collapse
Affiliation(s)
- Marine Queffeulou
- Centre de Recherche en Infectiologie du Centre de Recherche du CHU Québec, Université Laval, Québec, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, Canada
| | - Philippe Leprohon
- Centre de Recherche en Infectiologie du Centre de Recherche du CHU Québec, Université Laval, Québec, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, Canada
| | - Christopher Fernandez-Prada
- Centre de Recherche en Infectiologie du Centre de Recherche du CHU Québec, Université Laval, Québec, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, Canada
| | - Marc Ouellette
- Centre de Recherche en Infectiologie du Centre de Recherche du CHU Québec, Université Laval, Québec, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, Canada
| | - Ana María Mejía-Jaramillo
- Centre de Recherche en Infectiologie du Centre de Recherche du CHU Québec, Université Laval, Québec, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, Canada
| |
Collapse
|
5
|
Burge RJ, Jameson KH, Geoghegan V, Dowle AA, Mottram JC, Wilkinson AJ. Formation of functional E3 ligase complexes with UBC2 and UEV1 of Leishmania mexicana. Mol Biochem Parasitol 2024; 258:111619. [PMID: 38556171 DOI: 10.1016/j.molbiopara.2024.111619] [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: 01/08/2024] [Revised: 03/14/2024] [Accepted: 03/26/2024] [Indexed: 04/02/2024]
Abstract
In eukaryotic cells, molecular fate and cellular responses are shaped by multicomponent enzyme systems which reversibly attach ubiquitin and ubiquitin-like modifiers to target proteins. The extent of the ubiquitin proteasome system in Leishmania mexicana and its importance for parasite survival has recently been established through deletion mutagenesis and life-cycle phenotyping studies. The ubiquitin conjugating E2 enzyme UBC2, and the E2 enzyme variant UEV1, with which it forms a stable complex in vitro, were shown to be essential for the differentiation of promastigote parasites to the infectious amastigote form. To investigate further, we used immunoprecipitation of Myc-UBC2 or Myc-UEV1 to identify interacting proteins in L. mexicana promastigotes. The interactome of UBC2 comprises multiple ubiquitin-proteasome components including UEV1 and four RING E3 ligases, as well as potential substrates predicted to have roles in carbohydrate metabolism and intracellular trafficking. The smaller UEV1 interactome comprises six proteins, including UBC2 and shared components of the UBC2 interactome consistent with the presence of intracellular UBC2-UEV1 complexes. Recombinant RING1, RING2 and RING4 E3 ligases were shown to support ubiquitin transfer reactions involving the E1, UBA1a, and UBC2 to available substrate proteins or to unanchored ubiquitin chains. These studies define additional components of a UBC2-dependent ubiquitination pathway shown previously to be essential for promastigote to amastigote differentiation.
Collapse
Affiliation(s)
- Rebecca J Burge
- York Biomedical Research Institute, Department of Biology, University of York, York YO10 5DD, UK
| | - Katie H Jameson
- York Structural Biology Laboratory and York Biomedical Research Institute, Department of Chemistry, University of York, York YO10 5DD, UK
| | - Vincent Geoghegan
- York Biomedical Research Institute, Department of Biology, University of York, York YO10 5DD, UK
| | - Adam A Dowle
- Bioscience Technology Facility, Department of Biology, University of York, York YO10 5DD, UK
| | - Jeremy C Mottram
- York Biomedical Research Institute, Department of Biology, University of York, York YO10 5DD, UK.
| | - Anthony J Wilkinson
- York Structural Biology Laboratory and York Biomedical Research Institute, Department of Chemistry, University of York, York YO10 5DD, UK.
| |
Collapse
|
6
|
Marín M, López M, Gallego-Yerga L, Álvarez R, Peláez R. Experimental structure based drug design (SBDD) applications for anti-leishmanial drugs: A paradigm shift? Med Res Rev 2024; 44:1055-1120. [PMID: 38142308 DOI: 10.1002/med.22005] [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: 05/04/2023] [Revised: 11/14/2023] [Accepted: 11/27/2023] [Indexed: 12/25/2023]
Abstract
Leishmaniasis is a group of neglected tropical diseases caused by at least 20 species of Leishmania protozoa, which are spread by the bite of infected sandflies. There are three main forms of the disease: cutaneous leishmaniasis (CL, the most common), visceral leishmaniasis (VL, also known as kala-azar, the most serious), and mucocutaneous leishmaniasis. One billion people live in areas endemic to leishmaniasis, with an annual estimation of 30,000 new cases of VL and more than 1 million of CL. New treatments for leishmaniasis are an urgent need, as the existing ones are inefficient, toxic, and/or expensive. We have revised the experimental structure-based drug design (SBDD) efforts applied to the discovery of new drugs against leishmaniasis. We have grouped the explored targets according to the metabolic pathways they belong to, and the key achieved advances are highlighted and evaluated. In most cases, SBDD studies follow high-throughput screening campaigns and are secondary to pharmacokinetic optimization, due to the majoritarian belief that there are few validated targets for SBDD in leishmaniasis. However, some SBDD strategies have significantly contributed to new drug candidates against leishmaniasis and a bigger number holds promise for future development.
Collapse
Affiliation(s)
- Miguel Marín
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Marta López
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Laura Gallego-Yerga
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Raquel Álvarez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| | - Rafael Peláez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, Salamanca, Spain
| |
Collapse
|
7
|
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
|
8
|
Thomas M, McGonagle K, Rowland P, Robinson DA, Dodd PG, Camino-Díaz I, Campbell L, Cantizani J, Castañeda P, Conn D, Craggs PD, Edwards D, Ferguson L, Fosberry A, Frame L, Goswami P, Hu X, Korczynska J, MacLean L, Martin J, Mutter N, Osuna-Cabello M, Paterson C, Peña I, Pinto EG, Pont C, Riley J, Shishikura Y, Simeons FRC, Stojanovski L, Thomas J, Wrobel K, Young RJ, Zmuda F, Zuccotto F, Read KD, Gilbert IH, Marco M, Miles TJ, Manzano P, De Rycker M. Structure-Guided Design and Synthesis of a Pyridazinone Series of Trypanosoma cruzi Proteasome Inhibitors. J Med Chem 2023; 66:10413-10431. [PMID: 37506194 PMCID: PMC10424187 DOI: 10.1021/acs.jmedchem.3c00582] [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] [Received: 03/31/2023] [Indexed: 07/30/2023]
Abstract
There is an urgent need for new treatments for Chagas disease, a parasitic infection which mostly impacts South and Central America. We previously reported on the discovery of GSK3494245/DDD01305143, a preclinical candidate for visceral leishmaniasis which acted through inhibition of the Leishmania proteasome. A related analogue, active against Trypanosoma cruzi, showed suboptimal efficacy in an animal model of Chagas disease, so alternative proteasome inhibitors were investigated. Screening a library of phenotypically active analogues against the T. cruzi proteasome identified an active, selective pyridazinone, the development of which is described herein. We obtained a cryo-EM co-structure of proteasome and a key inhibitor and used this to drive optimization of the compounds. Alongside this, optimization of the absorption, distribution, metabolism, and excretion (ADME) properties afforded a suitable compound for mouse efficacy studies. The outcome of these studies is discussed, alongside future plans to further understand the series and its potential to deliver a new treatment for Chagas disease.
Collapse
Affiliation(s)
- Michael
G. Thomas
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Kate McGonagle
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Paul Rowland
- GlaxoSmithKline,
Chemistry, Medicines Research Centre, Gunnels Wood Road, Stevenage, U.K., SG1 2NY
| | - David A. Robinson
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Peter G. Dodd
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Isabel Camino-Díaz
- GlaxoSmithKline,
Discovery DMPK, IVIVT, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Lorna Campbell
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Juan Cantizani
- GlaxoSmithKline,
Global Health R&D, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Pablo Castañeda
- GlaxoSmithKline,
Discovery DMPK, IVIVT, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Daniel Conn
- GlaxoSmithKline,
Chemistry, Medicines Research Centre, Gunnels Wood Road, Stevenage, U.K., SG1 2NY
| | - Peter D. Craggs
- GlaxoSmithKline,
Chemistry, Medicines Research Centre, Gunnels Wood Road, Stevenage, U.K., SG1 2NY
| | - Darren Edwards
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Liam Ferguson
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Andrew Fosberry
- GlaxoSmithKline,
Chemistry, Medicines Research Centre, Gunnels Wood Road, Stevenage, U.K., SG1 2NY
| | - Laura Frame
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Panchali Goswami
- GlaxoSmithKline,
Chemistry, Medicines Research Centre, Gunnels Wood Road, Stevenage, U.K., SG1 2NY
| | - Xiao Hu
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Justyna Korczynska
- GlaxoSmithKline,
Chemistry, Medicines Research Centre, Gunnels Wood Road, Stevenage, U.K., SG1 2NY
| | - Lorna MacLean
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Julio Martin
- GlaxoSmithKline,
Global Health R&D, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Nicole Mutter
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Maria Osuna-Cabello
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Christy Paterson
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Imanol Peña
- GlaxoSmithKline,
Global Health R&D, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Erika G. Pinto
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Caterina Pont
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Jennifer Riley
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Yoko Shishikura
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Frederick R. C. Simeons
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Laste Stojanovski
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - John Thomas
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Karolina Wrobel
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | | | - Filip Zmuda
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Fabio Zuccotto
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Kevin D. Read
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Ian H. Gilbert
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| | - Maria Marco
- GlaxoSmithKline,
Global Health R&D, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Timothy J. Miles
- GlaxoSmithKline,
Global Health R&D, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Pilar Manzano
- GlaxoSmithKline,
Global Health R&D, Severo Ochoa 2, PTM, Tres Cantos, Madrid ES 28760, Spain
| | - Manu De Rycker
- Drug
Discovery Unit, University of Dundee, School
of Life Sciences, Dow Street, Dundee, U.K., DD1 5EH
| |
Collapse
|
9
|
Jamabo M, Mahlalela M, Edkins AL, Boshoff A. Tackling Sleeping Sickness: Current and Promising Therapeutics and Treatment Strategies. Int J Mol Sci 2023; 24:12529. [PMID: 37569903 PMCID: PMC10420020 DOI: 10.3390/ijms241512529] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Human African trypanosomiasis is a neglected tropical disease caused by the extracellular protozoan parasite Trypanosoma brucei, and targeted for eradication by 2030. The COVID-19 pandemic contributed to the lengthening of the proposed time frame for eliminating human African trypanosomiasis as control programs were interrupted. Armed with extensive antigenic variation and the depletion of the B cell population during an infectious cycle, attempts to develop a vaccine have remained unachievable. With the absence of a vaccine, control of the disease has relied heavily on intensive screening measures and the use of drugs. The chemotherapeutics previously available for disease management were plagued by issues such as toxicity, resistance, and difficulty in administration. The approval of the latest and first oral drug, fexinidazole, is a major chemotherapeutic achievement for the treatment of human African trypanosomiasis in the past few decades. Timely and accurate diagnosis is essential for effective treatment, while poor compliance and resistance remain outstanding challenges. Drug discovery is on-going, and herein we review the recent advances in anti-trypanosomal drug discovery, including novel potential drug targets. The numerous challenges associated with disease eradication will also be addressed.
Collapse
Affiliation(s)
- Miebaka Jamabo
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6139, South Africa; (M.J.); (M.M.)
| | - Maduma Mahlalela
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6139, South Africa; (M.J.); (M.M.)
| | - Adrienne L. Edkins
- Department of Biochemistry and Microbiology, Biomedical Biotechnology Research Centre (BioBRU), Rhodes University, Makhanda 6139, South Africa;
| | - Aileen Boshoff
- Biotechnology Innovation Centre, Rhodes University, Makhanda 6139, South Africa; (M.J.); (M.M.)
| |
Collapse
|
10
|
Danazumi AU, Ishmam IT, Idris S, Izert MA, Balogun EO, Górna MW. Targeted protein degradation might present a novel therapeutic approach in the fight against African trypanosomiasis. Eur J Pharm Sci 2023; 186:106451. [PMID: 37088149 PMCID: PMC11032742 DOI: 10.1016/j.ejps.2023.106451] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/11/2023] [Accepted: 04/20/2023] [Indexed: 04/25/2023]
Abstract
African trypanosomiasis (AT) is a hemoparasitic disease caused by infection with African trypanosomes and it is prevalent in many sub-Saharan African countries, affecting both humans and domestic animals. The disease is transmitted mostly by haematophagous insects of the genus Glossina while taking blood meal, in the process spreading the parasites from an infected animal to an uninfected animal. The disease is fatal if untreated, and the available drugs are generally ineffective and resulting in toxicities. Therefore, it is still pertinent to explore novel methods and targets for drug discovery. Proteolysis-targeting chimeras (PROTACs) present a new strategy for development of therapeutic molecules that mimic cellular proteasomal-mediated protein degradation to target proteins involved in different disease types. PROTACs have been used to degrade proteins involved in various cancers, neurodegenerative diseases, and immune disorders with remarkable success. Here, we highlight the problems associated with the current treatments for AT, discuss the concept of PROTACs and associated targeted protein degradation (TPD) approaches, and provide some insights on the future potential for the use of these emerging technologies (PROTACs and TPD) for the development of new generation of anti-Trypanosoma drugs and the first "TrypPROTACs".
Collapse
Affiliation(s)
- Ammar Usman Danazumi
- Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw, Warsaw, Poland; Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland; Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria
| | | | - Salisu Idris
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria
| | - Matylda Anna Izert
- Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw, Warsaw, Poland
| | - Emmanuel Oluwadare Balogun
- Department of Biochemistry, Ahmadu Bello University, Zaria, Nigeria; African Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University, Zaria, Nigeria.
| | - Maria Wiktoria Górna
- Biological and Chemical Research Centre, Department of Chemistry, University of Warsaw, Warsaw, Poland.
| |
Collapse
|
11
|
Gabaldón-Figueira JC, Martinez-Peinado N, Escabia E, Ros-Lucas A, Chatelain E, Scandale I, Gascon J, Pinazo MJ, Alonso-Padilla J. State-of-the-Art in the Drug Discovery Pathway for Chagas Disease: A Framework for Drug Development and Target Validation. Res Rep Trop Med 2023; 14:1-19. [PMID: 37337597 PMCID: PMC10277022 DOI: 10.2147/rrtm.s415273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/03/2023] [Indexed: 06/21/2023] Open
Abstract
Chagas disease is the most important protozoan infection in the Americas, and constitutes a significant public health concern throughout the world. Development of new medications against its etiologic agent, Trypanosoma cruzi, has been traditionally slow and difficult, lagging in comparison with diseases caused by other kinetoplastid parasites. Among the factors that explain this are the incompletely understood mechanisms of pathogenesis of T. cruzi infection and its complex set of interactions with the host in the chronic stage of the disease. These demand the performance of a variety of in vitro and in vivo assays as part of any drug development effort. In this review, we discuss recent breakthroughs in the understanding of the parasite's life cycle and their implications in the search for new chemotherapeutics. For this, we present a framework to guide drug discovery efforts against Chagas disease, considering state-of-the-art preclinical models and recently developed tools for the identification and validation of molecular targets.
Collapse
Affiliation(s)
| | - Nieves Martinez-Peinado
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
| | - Elisa Escabia
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
| | - Albert Ros-Lucas
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| | - Eric Chatelain
- Drugs for Neglected Diseases Initiative (DNDi), Geneva, Switzerland
| | - Ivan Scandale
- Drugs for Neglected Diseases Initiative (DNDi), Geneva, Switzerland
| | - Joaquim Gascon
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| | - María-Jesús Pinazo
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
- Drugs for Neglected Diseases Initiative (DNDi), Geneva, Switzerland
| | - Julio Alonso-Padilla
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic—University of Barcelona, Barcelona, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III (CIBERINFEC, ISCIII), Madrid, Spain
| |
Collapse
|
12
|
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
|
13
|
Caroli AP, Mansoldo FRP, Cardoso VS, Lage CLS, Carmo FL, Supuran CT, Beatriz Vermelho A. Are patents important indicators of innovation for Chagas disease treatment? Expert Opin Ther Pat 2023; 33:193-209. [PMID: 36786067 DOI: 10.1080/13543776.2023.2176219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
INTRODUCTION Chagas disease is a neglected, endemic disease in 21 countries, spreading to non-endemic countries too. Like other neglected diseases affecting primarily low- and middle-income countries, low investment and the absence of new chemical entities from the industry occurred. Increased knowledge about the parasite, drug targets, and vector control has been observed, but this was not translated into new drugs. The partnerships of pharmaceutical companies with academies and consolidated networks to increment the new drugs and treatment research in Chagas disease are shown. The current review analyzes in detail the patents dealing with compounds candidates for new drugs and treatment. The patent search was performed using Orbit Intelligence® software in the 2001-2021 period. AREAS COVERED The author focused specifically on patents for the treatment, the new candidates disclosed in the patents, and the barriers to innovation. EXPERT OPINION Patents in Chagas disease have been increasing in the last years, although they do not bring new compounds to an effective treatment.
Collapse
Affiliation(s)
- Andrea Pestana Caroli
- Federal University of Rio de Janeiro (UFRJ), Institute of Microbiology Paulo de Góes, BIOINOVAR - Biocatalysis, Bioproducts and Bioenergy, Rio de Janeiro, Brazil
| | - Felipe R P Mansoldo
- Federal University of Rio de Janeiro (UFRJ), Institute of Microbiology Paulo de Góes, BIOINOVAR - Biocatalysis, Bioproducts and Bioenergy, Rio de Janeiro, Brazil
| | - Veronica S Cardoso
- Federal University of Rio de Janeiro (UFRJ), Institute of Microbiology Paulo de Góes, BIOINOVAR - Biocatalysis, Bioproducts and Bioenergy, Rio de Janeiro, Brazil
| | - Celso Luiz Salgueiro Lage
- National Institute of Intellectual Property (INPI), Graduate and Research Division, Rio de Janeiro-RJ, Brazil
| | - Flavia L Carmo
- Federal University of Rio de Janeiro (UFRJ), Institute of Microbiology Paulo de Góes, LEMM - Molecular Microbial Ecology Laboratory
| | - Claudiu T Supuran
- NEUROFARBA Department Sezione di Scienze Farmaceutiche, Università degli Studi di Firenze, Sesto Fiorentino (Florence), Italy
| | - Alane Beatriz Vermelho
- Federal University of Rio de Janeiro (UFRJ), Institute of Microbiology Paulo de Góes, BIOINOVAR - Biocatalysis, Bioproducts and Bioenergy, Rio de Janeiro, Brazil
| |
Collapse
|
14
|
Nath SK, Pankajakshan P, Sharma T, Kumari P, Shinde S, Garg N, Mathur K, Arambam N, Harjani D, Raj M, Kwatra G, Venkatesh S, Choudhoury A, Bano S, Tayal P, Sharan M, Arora R, Strych U, Hotez PJ, Bottazzi ME, Rawal K. A Data-Driven Approach to Construct a Molecular Map of Trypanosoma cruzi to Identify Drugs and Vaccine Targets. Vaccines (Basel) 2023; 11:vaccines11020267. [PMID: 36851145 PMCID: PMC9963959 DOI: 10.3390/vaccines11020267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/28/2023] Open
Abstract
Chagas disease (CD) is endemic in large parts of Central and South America, as well as in Texas and the southern regions of the United States. Successful parasites, such as the causative agent of CD, Trypanosoma cruzi have adapted to specific hosts during their phylogenesis. In this work, we have assembled an interactive network of the complex relations that occur between molecules within T. cruzi. An expert curation strategy was combined with a text-mining approach to screen 10,234 full-length research articles and over 200,000 abstracts relevant to T. cruzi. We obtained a scale-free network consisting of 1055 nodes and 874 edges, and composed of 838 proteins, 43 genes, 20 complexes, 9 RNAs, 36 simple molecules, 81 phenotypes, and 37 known pharmaceuticals. Further, we deployed an automated docking pipeline to conduct large-scale docking studies involving several thousand drugs and potential targets to identify network-based binding propensities. These experiments have revealed that the existing FDA-approved drugs benznidazole (Bz) and nifurtimox (Nf) show comparatively high binding energies to the T. cruzi network proteins (e.g., PIF1 helicase-like protein, trans-sialidase), when compared with control datasets consisting of proteins from other pathogens. We envisage this work to be of value to those interested in finding new vaccines for CD, as well as drugs against the T. cruzi parasite.
Collapse
Affiliation(s)
- Swarsat Kaushik Nath
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Preeti Pankajakshan
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Trapti Sharma
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Priya Kumari
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Sweety Shinde
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Nikita Garg
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Kartavya Mathur
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Nevidita Arambam
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Divyank Harjani
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Manpriya Raj
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Garwit Kwatra
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Sayantan Venkatesh
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Alakto Choudhoury
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Saima Bano
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Prashansa Tayal
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Mahek Sharan
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Ruchika Arora
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
| | - Ulrich Strych
- Texas Children’s Hospital Center for Vaccine Development, Departments of Pediatrics and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Peter J. Hotez
- Texas Children’s Hospital Center for Vaccine Development, Departments of Pediatrics and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Biology, Baylor University, Waco, TX 76798, USA
| | - Maria Elena Bottazzi
- Texas Children’s Hospital Center for Vaccine Development, Departments of Pediatrics and Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Biology, Baylor University, Waco, TX 76798, USA
| | - Kamal Rawal
- Centre for Computational Biology and Bioinformatics, Amity Institute of Biotechnology, Amity University, Noida 201303, Uttar Pradesh, India
- Correspondence:
| |
Collapse
|
15
|
Comparative Proteomics and Genome-Wide Druggability Analyses Prioritized Promising Therapeutic Targets against Drug-Resistant Leishmania tropica. Microorganisms 2023; 11:microorganisms11010228. [PMID: 36677520 PMCID: PMC9860978 DOI: 10.3390/microorganisms11010228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/04/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Leishmania tropica is a tropical parasite causing cutaneous leishmaniasis (CL) in humans. Leishmaniasis is a serious public health threat, affecting an estimated 350 million people in 98 countries. The global rise in antileishmanial drug resistance has triggered the need to explore novel therapeutic strategies against this parasite. In the present study, we utilized the recently available multidrug resistant L. tropica strain proteome data repository to identify alternative therapeutic drug targets based on comparative subtractive proteomic and druggability analyses. Additionally, small drug-like compounds were scanned against novel targets based on virtual screening and ADME profiling. The analysis unveiled 496 essential cellular proteins of L. tropica that were nonhomologous to the human proteome set. The druggability analyses prioritized nine parasite-specific druggable proteins essential for the parasite's basic cellular survival, growth, and virulence. These prioritized proteins were identified to have appropriate binding pockets to anchor small drug-like compounds. Among these, UDPase and PCNA were prioritized as the top-ranked druggable proteins. The pharmacophore-based virtual screening and ADME profiling predicted MolPort-000-730-162 and MolPort-020-232-354 as the top hit drug-like compounds from the Pharmit resource to inhibit L. tropica UDPase and PCNA, respectively. The alternative drug targets and drug-like molecules predicted in the current study lay the groundwork for developing novel antileishmanial therapies.
Collapse
|
16
|
The Dynamics of Cryptococcus neoformans Cell and Transcriptional Remodeling during Infection. Cells 2022; 11:cells11233896. [PMID: 36497155 PMCID: PMC9740611 DOI: 10.3390/cells11233896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
The phenotypic plasticity of Cryptococcus neoformans is widely studied and demonstrated in vitro, but its influence on pathogenicity remains unclear. In this study, we investigated the dynamics of cryptococcal cell and transcriptional remodeling during pulmonary infection in a murine model. We showed that in Cryptococcus neoformans, cell size reduction (cell body ≤ 3 µm) is important for initial adaptation during infection. This change was associated with reproductive fitness and tissue invasion. Subsequently, the fungus develops mechanisms aimed at resistance to the host’s immune response, which is determinant for virulence. We investigated the transcriptional changes involved in this cellular remodeling and found an upregulation of transcripts related to ribosome biogenesis at the beginning (6 h) of infection and a later (10 days) upregulation of transcripts involved in the inositol pathway, energy production, and the proteasome. Consistent with a role for the proteasome, we found that its inhibition delayed cell remodeling during infection with the H99 strain. Altogether, these results further our understanding of the infection biology of C. neoformans and provide perspectives to support therapeutic and diagnostic targets for cryptococcosis.
Collapse
|
17
|
Sandes JM, de Figueiredo RCBQ. The endoplasmic reticulum of trypanosomatids: An unrevealed road for chemotherapy. Front Cell Infect Microbiol 2022; 12:1057774. [PMID: 36439218 PMCID: PMC9684732 DOI: 10.3389/fcimb.2022.1057774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 10/24/2022] [Indexed: 01/04/2024] Open
Abstract
The endoplasmic reticulum (ER) of higher eukaryotic cells forms an intricate membranous network that serves as the main processing facility for folding and assembling of secreted and membrane proteins. The ER is a highly dynamic organelle that interacts with other intracellular structures, as well as endosymbiotic pathogenic and non-pathogenic microorganisms. A strict ER quality control (ERQC) must work to ensure that proteins entering the ER are folded and processed correctly. Unfolded or misfolded proteins are usually identified, selected, and addressed to Endoplasmic Reticulum-Associated Degradation (ERAD) complex. Conversely, when there is a large demand for secreted proteins or ER imbalance, the accumulation of unfolded or misfolded proteins activates the Unfold Protein Response (UPR) to restore the ER homeostasis or, in the case of persistent ER stress, induces the cell death. Pathogenic trypanosomatids, such as Trypanosoma cruzi, Trypanosoma brucei and Leishmania spp are the etiological agents of important neglected diseases. These protozoans have a complex life cycle alternating between vertebrate and invertebrate hosts. The ER of trypanosomatids, like those found in higher eukaryotes, is also specialized for secretion, and depends on the ERAD and non-canonical UPR to deal with the ER stress. Here, we reviewed the basic aspects of ER biology, organization, and quality control in trypanosomatids. We also focused on the unusual way by which T. cruzi, T. brucei, and Leishmania spp. respond to ER stress, emphasizing how these parasites' ER-unrevealed roads might be an attractive target for chemotherapy.
Collapse
Affiliation(s)
- Jana Messias Sandes
- Laboratório de Biologia Celular e Molecular de Patógenos, Departamento de Microbiologia, Instituto Aggeu Magalhães, Recife, Brazil
- Laboratório de Microscopia Eletrônica, Instituto Keizo Assami, Universidade Federal de Pernambuco, Recife, Brazil
| | | |
Collapse
|
18
|
Protein Myristoylation Plays a Role in the Nuclear Entry of the Parvovirus Minute Virus of Mice. J Virol 2022; 96:e0111822. [PMID: 35950857 PMCID: PMC9472656 DOI: 10.1128/jvi.01118-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Being nonpathogenic to humans, rodent parvoviruses (PVs) are naturally oncolytic viruses with great potential as anti-cancer agents. As these viruses replicate in the host cell nucleus, they must gain access to the nucleus during infection. The PV minute virus of mice (MVM) and several other PVs transiently disrupt the nuclear envelope (NE) and enter the nucleus through the resulting breaks. However, the molecular basis of this unique nuclear entry pathway remains uncharacterized. In this study, we used MVM as a model to investigate the molecular mechanism by which PVs induce NE disruption during viral nuclear entry. By combining bioinformatics analyses, metabolic labeling assays, mutagenesis, and pharmacological inhibition, we identified a functional myristoylation site at the sequence 78GGKVGH83 of the unique portion of the capsid protein VP1 (VP1u) of MVM. Performing proteolytic cleavage studies with a peptide containing this myristoylation site or with purified virions, we found tryptophan at position 77 of MVM VP1u is susceptible to chymotrypsin cleavage, implying this cleavage exposes G (glycine) 78 at the N-terminus of VP1u for myristoylation. Subsequent experiments using inhibitors of myristoylation and cellular proteases with MVM-infected cells, or an imaging-based quantitative NE permeabilization assay, further indicate protein myristoylation and a chymotrypsin-like activity are essential for MVM to locally disrupt the NE during viral nuclear entry. We thus propose a model for the nuclear entry of MVM in which NE disruption is mediated by VP1u myristoylation after the intact capsid undergoes proteolytic processing to expose the required N-terminal G for myristoylation. IMPORTANCE Rodent parvoviruses (PVs), including minute virus of mice (MVM), have the ability to infect and kill cancer cells and thereby possess great potential in anti-cancer therapy. In fact, some of these viruses are currently being investigated in both preclinical studies and clinical trials to treat a wide variety of cancers. However, the detailed mechanism of how PVs enter the cell nucleus remains unknown. In this study, we for the first time demonstrated a chemical modification called "myristoylation" of a MVM protein plays an essential role in the nuclear entry of the virus. We also showed, in addition to protein myristoylation, a chymotrypsin-like activity, which may come from cellular proteasomes, is required for MVM to get myristoylated and enter the nucleus. These findings deepen our understanding on how MVM and other related PVs infect host cells and provide new insights for the development of PV-based anti-cancer therapies.
Collapse
|
19
|
Ibarra-Meneses AV, Corbeil A, Wagner V, Beaudry F, do Monte-Neto RL, Fernandez-Prada C. Exploring direct and indirect targets of current antileishmanial drugs using a novel thermal proteomics profiling approach. Front Cell Infect Microbiol 2022; 12:954144. [PMID: 35992178 PMCID: PMC9381709 DOI: 10.3389/fcimb.2022.954144] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
Visceral leishmaniasis (VL), caused by Leishmania infantum, is an oft-fatal neglected tropical disease. In the absence of an effective vaccine, the control of leishmaniasis relies exclusively on chemotherapy. Due to the lack of established molecular/genetic markers denoting parasite resistance, clinical treatment failure is often used as an indicator. Antimony-based drugs have been the standard antileishmanial treatment for more than seven decades, leading to major drug resistance in certain regions. Likewise, drug resistance to miltefosine and amphotericin B continues to spread at alarming rates. In consequence, innovative approaches are needed to accelerate the identification of antimicrobial drug targets and resistance mechanisms. To this end, we have implemented a novel approach based on thermal proteome profiling (TPP) to further characterize the mode of action of antileishmanials antimony, miltefosine and amphotericin B, as well as to better understand the mechanisms of drug resistance deployed by Leishmania. Proteins become more resistant to heat-induced denaturation when complexed with a ligand. In this way, we used multiplexed quantitative mass spectrometry-based proteomics to monitor the melting profile of thousands of expressed soluble proteins in WT, antimony-resistant, miltefosine-resistant, and amphotericin B-resistant L. infantum parasites, in the presence (or absence) of the above-mentioned drugs. Bioinformatics analyses were performed, including data normalization, melting profile fitting, and identification of proteins that underwent changes (fold change > 4) caused by complexation with a drug. With this unique approach, we were able to narrow down the regions of the L. infantum proteome that interact with antimony, miltefosine, and amphotericin B; validating previously-identified and unveiling novel drug targets. Moreover, analyses revealed candidate proteins potentially involved in drug resistance. Interestingly, we detected thermal proximity coaggregation for several proteins belonging to the same metabolic pathway (i.e., tryparedoxin peroxidase and aspartate aminotransferase in proteins exposed to antimony), highlighting the importance of these pathways. Collectively, our results could serve as a jumping-off point for the future development of innovative diagnostic tools for the detection and evaluation of antimicrobial-resistant Leishmania populations, as well as open the door for new on-target therapies.
Collapse
Affiliation(s)
- Ana Victoria Ibarra-Meneses
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
- The Research Group on Infectious Diseases in Production Animals (GREMIP), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Audrey Corbeil
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
- The Research Group on Infectious Diseases in Production Animals (GREMIP), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Victoria Wagner
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
- The Research Group on Infectious Diseases in Production Animals (GREMIP), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Francis Beaudry
- Département de Biomédecine, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
- Centre de recherche sur le cerveau et l’apprentissage (CIRCA), Université de Montréal, Montréal, QC, Canada
| | - Rubens L. do Monte-Neto
- Biotechnology Applied to Pathogens (BAP) - Instituto René Rachou – Fundação Oswaldo Cruz/Fiocruz Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Christopher Fernandez-Prada
- Département de Pathologie et Microbiologie, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
- The Research Group on Infectious Diseases in Production Animals (GREMIP), Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
- *Correspondence: Christopher Fernandez-Prada,
| |
Collapse
|
20
|
Durão R, Ramalhete C, Madureira AM, Mendes E, Duarte N. Plant Terpenoids as Hit Compounds against Trypanosomiasis. Pharmaceuticals (Basel) 2022; 15:ph15030340. [PMID: 35337138 PMCID: PMC8951850 DOI: 10.3390/ph15030340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/22/2022] [Accepted: 03/04/2022] [Indexed: 02/04/2023] Open
Abstract
Human African trypanosomiasis (sleeping sickness) and American trypanosomiasis (Chagas disease) are vector-borne neglected tropical diseases, caused by the protozoan parasites Trypanosoma brucei and Trypanosoma cruzi, respectively. These diseases were circumscribed to South American and African countries in the past. However, human migration, military interventions, and climate changes have had an important effect on their worldwide propagation, particularly Chagas disease. Currently, the treatment of trypanosomiasis is not ideal, becoming a challenge in poor populations with limited resources. Exploring natural products from higher plants remains a valuable approach to find new hits and enlarge the pipeline of new drugs against protozoal human infections. This review covers the recent studies (2016–2021) on plant terpenoids, and their semi-synthetic derivatives, which have shown promising in vitro and in vivo activities against Trypanosoma parasites.
Collapse
Affiliation(s)
- Raquel Durão
- Research Institute for Medicines (iMED.Ulisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (R.D.); (C.R.); (A.M.M.); (E.M.)
| | - Cátia Ramalhete
- Research Institute for Medicines (iMED.Ulisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (R.D.); (C.R.); (A.M.M.); (E.M.)
- ATLANTICA—Instituto Universitário, Fábrica da Pólvora de Barcarena, 2730-036 Barcarena, Portugal
| | - Ana Margarida Madureira
- Research Institute for Medicines (iMED.Ulisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (R.D.); (C.R.); (A.M.M.); (E.M.)
| | - Eduarda Mendes
- Research Institute for Medicines (iMED.Ulisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (R.D.); (C.R.); (A.M.M.); (E.M.)
| | - Noélia Duarte
- Research Institute for Medicines (iMED.Ulisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (R.D.); (C.R.); (A.M.M.); (E.M.)
- Correspondence:
| |
Collapse
|
21
|
Florin-Christensen M, Wieser SN, Suarez CE, Schnittger L. In Silico Survey and Characterization of Babesia microti Functional and Non-Functional Proteases. Pathogens 2021; 10:1457. [PMID: 34832610 PMCID: PMC8621943 DOI: 10.3390/pathogens10111457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/29/2021] [Accepted: 11/06/2021] [Indexed: 12/23/2022] Open
Abstract
Human babesiosis caused by the intraerythrocytic apicomplexan Babesia microti is an expanding tick-borne zoonotic disease that may cause severe symptoms and death in elderly or immunocompromised individuals. In light of an increasing resistance of B. microti to drugs, there is a lack of therapeutic alternatives. Species-specific proteases are essential for parasite survival and possible chemotherapeutic targets. However, the repertoire of proteases in B. microti remains poorly investigated. Herein, we employed several combined bioinformatics tools and strategies to organize and identify genes encoding for the full repertoire of proteases in the B. microti genome. We identified 64 active proteases and 25 nonactive protease homologs. These proteases can be classified into cysteine (n = 28), serine (n = 21), threonine (n = 14), asparagine (n = 7), and metallopeptidases (n = 19), which, in turn, are assigned to a total of 38 peptidase families. Comparative studies between the repertoire of B. bovis and B. microti proteases revealed differences among sensu stricto and sensu lato Babesia parasites that reflect their distinct evolutionary history. Overall, this data may help direct future research towards our understanding of the biology and pathogenicity of Babesia parasites and to explore proteases as targets for developing novel therapeutic interventions.
Collapse
Affiliation(s)
- Monica Florin-Christensen
- Instituto de Patobiologia Veterinaria (IPVET), Centro de Investigaciones en Ciencias Veterinarias y Agronomicas, Instituto Nacional de Tecnología Agropecuaria (INTA), Hurlingham C1033AAE, Argentina; (S.N.W.); (L.S.)
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1033AAJ, Argentina
| | - Sarah N. Wieser
- Instituto de Patobiologia Veterinaria (IPVET), Centro de Investigaciones en Ciencias Veterinarias y Agronomicas, Instituto Nacional de Tecnología Agropecuaria (INTA), Hurlingham C1033AAE, Argentina; (S.N.W.); (L.S.)
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1033AAJ, Argentina
| | - Carlos E. Suarez
- Animal Disease Research Unit, USDA-ARS, Pullman, WA 99163, USA;
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA 99163, USA
| | - Leonhard Schnittger
- Instituto de Patobiologia Veterinaria (IPVET), Centro de Investigaciones en Ciencias Veterinarias y Agronomicas, Instituto Nacional de Tecnología Agropecuaria (INTA), Hurlingham C1033AAE, Argentina; (S.N.W.); (L.S.)
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1033AAJ, Argentina
| |
Collapse
|
22
|
Pan K, Fu J, Xu W. Role of Ubiquitin-Specific Peptidase 47 in Cancers and Other Diseases. Front Cell Dev Biol 2021; 9:726632. [PMID: 34604226 PMCID: PMC8484750 DOI: 10.3389/fcell.2021.726632] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/24/2021] [Indexed: 12/24/2022] Open
Abstract
Deubiquitination is the reverse process of ubiquitination, which is catalyzed by deubiquitinase enzymes. More than 100 deubiquitinases have been identified. Ubiquitin-specific peptidase 47 (USP47), a member of the ubiquitin-specific protease family with high homology to USP7, is an active molecule with a wide range of functions and is closely associated with cancer and other diseases. However, no systematic summary exists regarding the functions of USP47. Here, we summarize the functions and expression regulation of USP47. USP47 is highly expressed in many tumors and is widely involved in tumor development, metastasis, drug resistance, epithelial-mesenchymal transition, and other processes. Targeted inhibition of USP47 can reverse malignant tumor behavior. USP47 also plays a role in inflammatory responses, myocardial infarction, and neuronal development. USP47 is involved in multiple levels of expression-regulating mechanisms, including transcriptional, post-transcriptional, and post-translational modifications. Development of targeted inhibitors against USP47 will provide a basis for studying the mechanisms of USP47 and developing therapeutic strategies for cancers and other diseases.
Collapse
Affiliation(s)
- Kailing Pan
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Junhao Fu
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Wenxia Xu
- Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|