1
|
Jackson V, Sherer C, Jordan L, Clohessy T. Unveiling the potential: exploring the efficacy of complex III inhibitors in fungal disease control. PEST MANAGEMENT SCIENCE 2025; 81:2450-2456. [PMID: 39177294 DOI: 10.1002/ps.8384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 08/24/2024]
Abstract
Phytopathogenic fungi are a key challenge to maximizing crop yield and quality for a growing global population. In this review, we give an overview of representative compounds that inhibit complex III, also known as bc1 complex, covering quinone inside inhibitors, quinone outside inhibitors, and quinone inside and outside inhibitors via the stigmatellin binding mode. Novel solutions to the escalating problem of resistance are still required, therefore compounds with alternative scaffolds, alternative docking modes, different mechanisms of action and improved efficacy against complex III necessitate ongoing research. © 2024 Society of Chemical Industry.
Collapse
|
2
|
Arantes GM. Redox-Activated Proton Transfer through a Redundant Network in the Q o Site of Cytochrome bc1. J Chem Inf Model 2025; 65:2660-2669. [PMID: 40008618 PMCID: PMC11898062 DOI: 10.1021/acs.jcim.4c02361] [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: 12/24/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 02/27/2025]
Abstract
Proton translocation catalyzed by cytochrome bc1 (respiratory complex III) during coenzyme-Q redox cycling is a critical bioenergetic process, yet its detailed molecular mechanism remains incompletely understood. In this study, the energetics of the long-range proton transfers through multiple proton-conducting wires in the Qo site of the bc1 complex was investigated computationally using hybrid QM/MM simulations and a specialized reaction coordinate. Key reactive groups and proton transfer mechanisms were characterized, confirming the propionate-A group of heme bL as a plausible proton acceptor. Upon coenzyme-Q oxidation, a Grotthuss hopping mechanism is activated, facilitating proton transfer along three distinct pathways with comparable barriers and stability. These pathways operate redundantly, forming a robust proton-conducting network, and account for the unusual experimental behavior observed in single-point mutations. Energetic analyses exclude charged closed-shell species as likely intermediates and propose a reaction sequence for coenzyme-Q oxidation proceeding as QH2 → QH• → Q0, either via coupled proton-electron transfers or stepwise mechanisms involving open-shell intermediates. These findings elucidate mechanistic details of the Q-cycle and improve our understanding of the catalytic reactions supporting redox-activated proton transfer in respiratory enzymes.
Collapse
Affiliation(s)
- Guilherme M. Arantes
- Department of Biochemistry,
Instituto de Química, Universidade
de São Paulo, Av. Prof. Lineu Prestes 748, 05508-900, São Paulo, SP, Brazil
| |
Collapse
|
3
|
Dong Y, Guan XY, Guan ZW, Zhang JC, Zhu XL, Yang GF. Triazole Sulfonamide Derivates: Inhibitors of the bc1 Complex to Control Cucumber Downy Mildew. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:27513-27520. [PMID: 39576844 DOI: 10.1021/acs.jafc.4c05904] [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: 11/24/2024]
Abstract
Cucumber downy mildew (CDM), caused by Pseudoperonospora cubensis, is a destructive disease that affects greenhouse cucumbers and causes significant losses for growers. Amisulbrom, a triazole sulfonamide fungicide targeting the Qi site in the bc1 complex, has shown potential in effectively combating CDM. However, its detailed binding mode with the target is unclear. In this study, a three-dimensional (3D) structure of the bc1 complex from P. cubensis was built, and its interaction with amisulbrom was investigated by integrating molecular docking, molecular dynamics, and molecular mechanics/Poisson-Boltzmann surface area (MM/PBSA) methods. Based on the binding mode of amisulbrom with the Pc-bc1 complex, a scaffold hopping strategy was performed, and compounds 11a-o and 12a-v were designed. Among them, compound 12g showed excellent fungicidal properties against CDM in field trials. The present work indicated that the oxime ether moiety could be further optimized for better results. Furthermore, compound 12g has the potential to serve as a lead compound in the search for new Qi-site inhibitors of the bc1 complex.
Collapse
Affiliation(s)
- Ying Dong
- State Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan 430079, PR China
| | - Xiao-Yan Guan
- State Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan 430079, PR China
| | - Ze-Wei Guan
- State Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan 430079, PR China
| | - Jun-Chao Zhang
- State Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan 430079, PR China
| | - Xiao-Lei Zhu
- State Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan 430079, PR China
| | - Guang-Fu Yang
- State Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensor Technology and Health, Central China Normal University, Wuhan 430079, PR China
- Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300071, People's Republic of China
| |
Collapse
|
4
|
Hewitt P, Hartmann A, Tornesi B, Ferry-Martin S, Valentin JP, Desert P, Gresham S, Demarta-Gatsi C, Venishetty VK, Kolly C. Importance of tailored non-clinical safety testing of novel antimalarial drugs: Industry best-practice. Regul Toxicol Pharmacol 2024; 154:105736. [PMID: 39515409 DOI: 10.1016/j.yrtph.2024.105736] [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: 08/09/2024] [Revised: 09/24/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Malaria is an acute, debilitating parasitic illness. There were 249 million cases of malaria in 2022, resulting in 608,000 deaths globally, 76% of which were children ≤5 years. The unique nature of this disease (recurrences leading to re-treatments and numerous organ systems affected), specific clinical treatment regimens, poor compliance, and diversity of affected populations (predominantly pediatrics, women of childbearing potential, pregnant and lactating women), often makes standard testing approaches inadequate, and tailor-made safety assessments are more appropriate. We provide best practice recommendations based on company experience for the non-clinical safety assessment of antimalarial drugs, with a focus on small molecules since they represent the majority of drug candidates for this illness. We focus on specific testing considerations for repeat dose toxicity studies, including combination toxicity assessments, since new drug treatment regimens typically foresee short treatment durations to improve compliance (i.e., 1 day) with combinations of compounds to improve efficacy and limit potential resistance. Due to the target population, the timing of reproductive, developmental, and juvenile toxicity studies may be earlier than general testing roadmaps for other small molecule drugs. In conclusion, key recommendations presented should enable a more effective and efficient development path whilst protecting clinical trial participants and patients.
Collapse
Affiliation(s)
- Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany.
| | | | - Belen Tornesi
- Non-Clinical Pharmacology & Toxicology, Medicines for Malaria Venture, Geneva, Switzerland
| | - Sandrine Ferry-Martin
- Nonclinical Drug Safety, Merck Research Laboratories, Merck Sharp & Dohme, Clermont-Ferrand, France
| | - Jean-Pierre Valentin
- Early Clinical Development & Translational Science, Non-Clinical Safety Evaluation, UCB Pharma, Braine L'Alleud, Belgium
| | - Paul Desert
- Nonclinical Safety, Sanofi, Marcy l'Etoile, France
| | | | - Claudia Demarta-Gatsi
- Global Health R&D of Merck Healthcare, Ares Trading S.A., (a subsidiary of Merck KGaA, Darmstadt, Germany), Switzerland
| | | | - Carine Kolly
- Preclinical Safety, Novartis Pharma AG, Basel, Switzerland
| |
Collapse
|
5
|
Cardillo NM, Villarino NF, Lacy PA, Riscoe MK, Doggett JS, Ueti MW, Chung CJ, Suarez CE. The Combination of Buparvaquone and ELQ316 Exhibit a Stronger Effect than ELQ316 and Imidocarb Against Babesia bovis In Vitro. Pharmaceutics 2024; 16:1402. [PMID: 39598526 PMCID: PMC11597495 DOI: 10.3390/pharmaceutics16111402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Bovine babesiosis is a vector-borne disease transmitted by ticks that causes important losses in livestock worldwide. Recent research performed on the drugs currently used to control bovine babesiosis reported several issues including drug resistance, toxicity impact, and residues in edible tissue, suggesting the need for developing novel effective therapies. The endochin-like quinolones ELQ-316 and buparvaquone (BPQ) act as cytochrome bc1 inhibitors and have been proven to be safe and efficacious against related apicomplexans, such as Plasmodium spp. and Babesia microti, without showing toxicity in mammals. The objectives of this study are investigating whether ELQ-316, BPQ, and their combination treatment could be effective against Babesia bovis in an in vitro culture model and comparing with imidocarb (ID), the routinely used drug. Methods: In vitro cultured parasites starting at 2% percentage of parasitemia (PPE) were treated with BPQ, ELQ-316, ID, and the combinations of BPQ + ELQ-316 and ID + ELQ-316 at drug concentrations that ranged from 25 to 1200 nM, during four consecutive days. The IC50% and IC99% were reported. Parasitemia levels were evaluated daily using microscopic examination. Data were compared using the non-parametrical Mann-Whitney and Kruskall-Wallis test. Results: All drugs tested, whether used alone or in combination, significantly decreased the survival (p < 0.05) of B. bovis in in vitro cultures. The combination of BPQ + ELQ-316 had the lowest calculated inhibitory concentration 50% (IC50%) values, 31.21 nM (IC95%: 15.06-68.48); followed by BPQ, 77.06 nM (IC95%: 70.16-86.01); ID + ELQ316, 197 nM (IC95%:129.0-311.2); ID, 635.1 nM (IC95%: 280.9-2119); and ELQ316, 654.9 nM (IC95%: 362.3-1411). Conclusions: The results reinforce the higher efficacy of BPQ at affecting B. bovis survival and the potential synergistic effects of its combination with ELQ-316, providing a promising treatment option against B. bovis.
Collapse
Affiliation(s)
- Natalia M. Cardillo
- Animal Disease Research Unit, United States Department of Agriculture-Animal Research Unit (USDA-ARS), 3003 ADBF, WSU, Pullman, WA 99163, USA; (P.A.L.); (M.W.U.); (C.J.C.); (C.E.S.)
- Estación Experimental INTA Paraná, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires 2290, Argentina
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA 99164, USA
| | - Nicolas F. Villarino
- Program in Individualized Medicine, Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA;
| | - Paul A. Lacy
- Animal Disease Research Unit, United States Department of Agriculture-Animal Research Unit (USDA-ARS), 3003 ADBF, WSU, Pullman, WA 99163, USA; (P.A.L.); (M.W.U.); (C.J.C.); (C.E.S.)
| | - Michael K. Riscoe
- VA Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, OR 97239, USA (J.S.D.)
- Department of Microbiology and Molecular Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Joseph Stone Doggett
- VA Portland Healthcare System, 3710 SW US Veterans Hospital Road, Portland, OR 97239, USA (J.S.D.)
- School of Medicine, Division of Infectious Diseases, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Massaro W. Ueti
- Animal Disease Research Unit, United States Department of Agriculture-Animal Research Unit (USDA-ARS), 3003 ADBF, WSU, Pullman, WA 99163, USA; (P.A.L.); (M.W.U.); (C.J.C.); (C.E.S.)
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA 99164, USA
| | - Chungwon J. Chung
- Animal Disease Research Unit, United States Department of Agriculture-Animal Research Unit (USDA-ARS), 3003 ADBF, WSU, Pullman, WA 99163, USA; (P.A.L.); (M.W.U.); (C.J.C.); (C.E.S.)
| | - Carlos E. Suarez
- Animal Disease Research Unit, United States Department of Agriculture-Animal Research Unit (USDA-ARS), 3003 ADBF, WSU, Pullman, WA 99163, USA; (P.A.L.); (M.W.U.); (C.J.C.); (C.E.S.)
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
6
|
Azevedo LG, Sosa E, de Queiroz ATL, Barral A, Wheeler RJ, Nicolás MF, Farias LP, Do Porto DF, Ramos PIP. High-throughput prioritization of target proteins for development of new antileishmanial compounds. Int J Parasitol Drugs Drug Resist 2024; 25:100538. [PMID: 38669848 PMCID: PMC11068527 DOI: 10.1016/j.ijpddr.2024.100538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 03/11/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024]
Abstract
Leishmaniasis, a vector-borne disease, is caused by the infection of Leishmania spp., obligate intracellular protozoan parasites. Presently, human vaccines are unavailable, and the primary treatment relies heavily on systemic drugs, often presenting with suboptimal formulations and substantial toxicity, making new drugs a high priority for LMIC countries burdened by the disease, but a low priority in the agenda of most pharmaceutical companies due to unattractive profit margins. New ways to accelerate the discovery of new, or the repositioning of existing drugs, are needed. To address this challenge, our study aimed to identify potential protein targets shared among clinically-relevant Leishmania species. We employed a subtractive proteomics and comparative genomics approach, integrating high-throughput multi-omics data to classify these targets based on different druggability metrics. This effort resulted in the ranking of 6502 ortholog groups of protein targets across 14 pathogenic Leishmania species. Among the top 20 highly ranked groups, metabolic processes known to be attractive drug targets, including the ubiquitination pathway, aminoacyl-tRNA synthetases, and purine synthesis, were rediscovered. Additionally, we unveiled novel promising targets such as the nicotinate phosphoribosyltransferase enzyme and dihydrolipoamide succinyltransferases. These groups exhibited appealing druggability features, including less than 40% sequence identity to the human host proteome, predicted essentiality, structural classification as highly druggable or druggable, and expression levels above the 50th percentile in the amastigote form. The resources presented in this work also represent a comprehensive collection of integrated data regarding trypanosomatid biology.
Collapse
Affiliation(s)
- Lucas G Azevedo
- Center for Data and Knowledge Integration for Health (CIDACS), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (Fiocruz Bahia), Salvador, Bahia, Brazil; Post-graduate Program in Biotechnology and Investigative Medicine, Instituto Gonçalo Moniz, Salvador, Bahia, Brazil.
| | - Ezequiel Sosa
- Universidad de Buenos Aires, Buenos Aires, Argentina.
| | - Artur T L de Queiroz
- Center for Data and Knowledge Integration for Health (CIDACS), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (Fiocruz Bahia), Salvador, Bahia, Brazil; Post-graduate Program in Biotechnology and Investigative Medicine, Instituto Gonçalo Moniz, Salvador, Bahia, Brazil.
| | - Aldina Barral
- Laboratório de Medicina e Saúde Pública de Precisão (MeSP2), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (Fiocruz Bahia), Salvador, Bahia, Brazil.
| | - Richard J Wheeler
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
| | - Marisa F Nicolás
- Laboratório Nacional de Computação Científica, Petrópolis, Rio de Janeiro, Brazil.
| | - Leonardo P Farias
- Post-graduate Program in Biotechnology and Investigative Medicine, Instituto Gonçalo Moniz, Salvador, Bahia, Brazil; Laboratório de Medicina e Saúde Pública de Precisão (MeSP2), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (Fiocruz Bahia), Salvador, Bahia, Brazil.
| | | | - Pablo Ivan P Ramos
- Center for Data and Knowledge Integration for Health (CIDACS), Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (Fiocruz Bahia), Salvador, Bahia, Brazil; Post-graduate Program in Biotechnology and Investigative Medicine, Instituto Gonçalo Moniz, Salvador, Bahia, Brazil.
| |
Collapse
|
7
|
Saldivia M, Lima APCA, Mottram JC. A promising pipeline of preclinical drug candidates for leishmaniasis and chronic Chagas' disease. Trends Parasitol 2024; 40:211-213. [PMID: 38368155 DOI: 10.1016/j.pt.2024.02.002] [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: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 02/19/2024]
Abstract
The drug discovery pipeline for leishmaniasis and trypanosomiasis has been filling with novel chemical entities with known mechanisms of action. González et al. and Braillard et al. report a cytochrome bc1 complex inhibitor as another promising preclinical candidate for visceral leishmaniasis (VL) and, in combination with benznidazole, for chronic Chagas' disease (CCD).
Collapse
Affiliation(s)
| | - Ana Paula C A Lima
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jeremy C Mottram
- York Biomedical Research Institute and Department of Biology, University of York, York, UK.
| |
Collapse
|
8
|
Dong Y, Li B, Yin MX, Liu Z, Niu Y, Wu QY, Zhu XL, Yang GF. The Interaction Mechanism of Picolinamide Fungicide Targeting on the Cytochrome bc1 Complex and Its Structural Modification. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:3755-3762. [PMID: 38346446 DOI: 10.1021/acs.jafc.3c05982] [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: 02/22/2024]
Abstract
Picolinamide fungicides, structurally related to UK-2A and antimycin-A, bind into the Qi-site in the bc1 complex. However, the detailed binding mode of picolinamide fungicides remains unknown. In the present study, antimycin-A and UK-2A were selected to study the binding mode of picolinamide inhibitors with four protonation states in the Qi-site by integrating molecular dynamics simulation, molecular docking, and molecular mechanics Generalized Born surface area (MM/GBSA) calculations. Subsequently, a series of new picolinamide derivatives were designed and synthesized to further understand the effects of substituents on the tail phenyl ring. The computational results indicated that the substituted aromatic rings in antimycin-A and UK-2A were the pharmacophore fragments and made the primary contribution when bound to a protein. Compound 9g-hydrolysis formed H-bonds with Hie201 and Ash228 and showed an IC50 value of 6.05 ± 0.24 μM against the porcine bc1 complex. Compound 9c, with a simpler chemical structure, showed higher control effects than florylpicoxamid against cucumber downy mildew and expanded the fungicidal spectrum of picolinamide fungicides. The structural and mechanistic insights obtained from the present study will provide a valuable clue for the future designing of new promising Qi-site inhibitors.
Collapse
Affiliation(s)
- Ying Dong
- National Key Laboratory of Green Pesticide, Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health of Ministry of Science and Technology, Central China Normal University, Wuhan 430079, People's Republic of China
| | - Bo Li
- National Key Laboratory of Green Pesticide, Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health of Ministry of Science and Technology, Central China Normal University, Wuhan 430079, People's Republic of China
| | - Mao-Xue Yin
- National Key Laboratory of Green Pesticide, Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health of Ministry of Science and Technology, Central China Normal University, Wuhan 430079, People's Republic of China
| | - Zheng Liu
- National Key Laboratory of Green Pesticide, Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health of Ministry of Science and Technology, Central China Normal University, Wuhan 430079, People's Republic of China
| | - Yan Niu
- National Key Laboratory of Green Pesticide, Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health of Ministry of Science and Technology, Central China Normal University, Wuhan 430079, People's Republic of China
| | - Qiong-You Wu
- National Key Laboratory of Green Pesticide, Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health of Ministry of Science and Technology, Central China Normal University, Wuhan 430079, People's Republic of China
| | - Xiao-Lei Zhu
- National Key Laboratory of Green Pesticide, Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health of Ministry of Science and Technology, Central China Normal University, Wuhan 430079, People's Republic of China
| | - Guang-Fu Yang
- National Key Laboratory of Green Pesticide, Key Laboratory of Pesticide & Chemical Biology of Ministry of Education, International Joint Research Center for Intelligent Biosensor Technology and Health of Ministry of Science and Technology, Central China Normal University, Wuhan 430079, People's Republic of China
- Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300071, People's Republic of China
| |
Collapse
|
9
|
Yolanda H, Jearawuttanakul K, Wannalo W, Kanjanasirirat P, Borwornpinyo S, Rujirawat T, Payattikul P, Kittichotirat W, Wichadakul D, Krajaejun T. Potential anti- Pythium insidiosum therapeutics identified through screening of agricultural fungicides. Microbiol Spectr 2024; 12:e0162023. [PMID: 38179943 PMCID: PMC10846074 DOI: 10.1128/spectrum.01620-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/07/2023] [Indexed: 01/06/2024] Open
Abstract
Pythiosis is a life-threatening infectious disease caused by the oomycete Pythium insidiosum. Clinical manifestations of pythiosis include an eye, blood vessel, skin, or gastrointestinal tract infection. Pythiosis has been increasingly reported worldwide, with an overall mortality rate of 28%. Radical surgery is required to save patients' lives due to the limited efficacy of antimicrobial drugs. Effective medical treatments are urgently needed for pythiosis. This study aims to find anti-P. insidiosum agents by screening 17 agricultural fungicides that inhibit plant-pathogenic oomycetes and validating their efficacy and safety. Cyazofamid outperformed other fungicides as it can potently inhibit genetically diverse P. insidiosum isolates while exhibiting minimal cellular toxicities. The calculated therapeutic scores determined that the concentration of cyazofamid causing significant cellular toxicities was eight times greater than the concentration of the drug effectively inhibiting P. insidiosum. Furthermore, other studies showed that cyazofamid exhibits low-to-moderate toxicities in animals. The mechanism of cyazofamid action is likely the inhibition of cytochrome b, an essential component in ATP synthesis. Molecular docking and dynamic analyses depicted a stable binding of cyazofamid to the Qi site of the P. insidiosum's cytochrome b orthologous protein. In conclusion, our search for an effective anti-P. insidiosum drug indicated that cyazofamid is a promising candidate for treating pythiosis. With its high efficacy and low toxicity, cyazofamid is a potential chemical for treating pythiosis, reducing the need for radical surgeries, and improving recovery rates. Our findings could pave the way for the development of new and effective treatments for pythiosis.IMPORTANCEPythiosis is a severe infection caused by Pythium insidiosum. The disease is prevalent in tropical/subtropical regions. This infectious condition is challenging to treat with antifungal drugs and often requires surgical removal of the infected tissue. Pythiosis can be fatal if not treated promptly. There is a need for a new treatment that effectively inhibits P. insidiosum. This study screened 17 agricultural fungicides that target plant-pathogenic oomycetes and found that cyazofamid was the most potent in inhibiting P. insidiosum. Cyazofamid showed low toxicity to mammalian cells and high affinity to the P. insidiosum's cytochrome b, which is involved in energy production. Cyazofamid could be a promising candidate for the treatment of pythiosis, as it could reduce the need for surgery and improve the survival rate of patients. This study provides valuable insights into the biology and drug susceptibility of P. insidiosum and opens new avenues for developing effective therapies for pythiosis.
Collapse
Affiliation(s)
- Hanna Yolanda
- Program in Translational Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
- Department of Parasitology, School of Medicine and Health Sciences, Atma Jaya Catholic University of Indonesia, Jakarta, Indonesia
| | - Kedchin Jearawuttanakul
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Warawuth Wannalo
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Suparerk Borwornpinyo
- Excellent Center for Drug Discovery, Faculty of Science, Mahidol University, Bangkok, Thailand
- Department of Biotechnology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Thidarat Rujirawat
- Research Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Penpan Payattikul
- Research Center, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Weerayuth Kittichotirat
- Bioinformatics and Systems Biology Program, School of Bioresources and Technology and School of Information Technology, King Mongkut’s University of Technology Thonburi, Bangkhuntien, Bangkok, Thailand
- Systems Biology and Bioinformatics Research Group, Pilot Plant Development and Training Institute, King Mongkut’s University of Technology Thonburi, Bangkhuntien, Bangkok, Thailand
| | - Duangdao Wichadakul
- Department of Computer Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Theerapong Krajaejun
- Department of Pathology, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
10
|
Wang Y, Yang J, Zhang Y, Zhou J. Focus on Mitochondrial Respiratory Chain: Potential Therapeutic Target for Chronic Renal Failure. Int J Mol Sci 2024; 25:949. [PMID: 38256023 PMCID: PMC10815764 DOI: 10.3390/ijms25020949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/26/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
The function of the respiratory chain is closely associated with kidney function, and the dysfunction of the respiratory chain is a primary pathophysiological change in chronic kidney failure. The incidence of chronic kidney failure caused by defects in respiratory-chain-related genes has frequently been overlooked. Correcting abnormal metabolic reprogramming, rescuing the "toxic respiratory chain", and targeting the clearance of mitochondrial reactive oxygen species are potential therapies for treating chronic kidney failure. These treatments have shown promising results in slowing fibrosis and inflammation progression and improving kidney function in various animal models of chronic kidney failure and patients with chronic kidney disease (CKD). The mitochondrial respiratory chain is a key target worthy of attention in the treatment of chronic kidney failure. This review integrated research related to the mitochondrial respiratory chain and chronic kidney failure, primarily elucidating the pathological status of the mitochondrial respiratory chain in chronic kidney failure and potential therapeutic drugs. It provided new ideas for the treatment of kidney failure and promoted the development of drugs targeting the mitochondrial respiratory chain.
Collapse
Affiliation(s)
| | | | | | - Jianhua Zhou
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China; (Y.W.); (J.Y.); (Y.Z.)
| |
Collapse
|
11
|
Zhang J, Gelain J, Schnabel G, Mallawarachchi S, Wang H, Mulgaonkar N, Karthikeyan R, Fernando S. Identification of Fungicide Combinations for Overcoming Plasmopara viticola and Botrytis cinerea Fungicide Resistance. Microorganisms 2023; 11:2966. [PMID: 38138109 PMCID: PMC10746041 DOI: 10.3390/microorganisms11122966] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/21/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Fungal diseases, including downy mildew (caused by Plasmopara viticola) and gray mold (caused by Botrytis cinerea), significantly impact the marketable yield of grapes produced worldwide. Cytochrome b of the mitochondrial respiratory chain of these two fungi is a key target for Quinone outside inhibitor (QoI)-based fungicide development. Since the mode of action (MOA) of QoI fungicides is restricted to a single site, the extensive usage of these fungicides has resulted in fungicide resistance. The use of fungicide combinations with multiple targets is an effective way to counter and slow down the development of fungicide resistance. Due to the high cost of in planta trials, in silico techniques can be used for the rapid screening of potential fungicides. In this study, a combination of in silico simulations that include Schrödinger Glide docking, molecular dynamics, and Molecular Mechanism-Generalized Born Surface Area calculation were used to screen the most potent QoI and non-QoI-based fungicide combinations to wild-type, G143A-mutated, F129L-mutated, and double-mutated versions that had both G143A and F129L mutations of fungal cytochrome b. In silico docking studies indicated that mandestrobin, famoxadone, captan, and thiram have a high affinity toward WT cytochrome b of Botrytis cinerea. Although the QoIs mandestrobin and famoxadone were effective for WT based on in vitro results, they were not broadly effective against G143A-mutated isolates. Famoxadone was only effective against one isolate with G143A-mutated cytochrome b. The non-QoI fungicides thiram and captan were effective against both WT and isolates with G143A-mutated cytochrome b. Follow-up in silico docking and molecular dynamics studies suggested that fungicide combinations consisting of famoxadone, mandestrobin, fenamidone, and thiram should be considered in field testing targeting Plasmopara viticola and Botrytis cinerea fungicide resistance.
Collapse
Affiliation(s)
- Junrui Zhang
- Department of Biological and Agricultural Engineering, Texas A&M University, College Station, TX 77845, USA
| | - Jhulia Gelain
- Department of Plant and Environmental Sciences, Clemson University, Clemson, SC 29634, USA
| | - Guido Schnabel
- Department of Plant and Environmental Sciences, Clemson University, Clemson, SC 29634, USA
| | - Samavath Mallawarachchi
- Department of Biological and Agricultural Engineering, Texas A&M University, College Station, TX 77845, USA
| | - Haoqi Wang
- Department of Biological and Agricultural Engineering, Texas A&M University, College Station, TX 77845, USA
| | - Nirmitee Mulgaonkar
- Department of Biological and Agricultural Engineering, Texas A&M University, College Station, TX 77845, USA
| | | | - Sandun Fernando
- Department of Biological and Agricultural Engineering, Texas A&M University, College Station, TX 77845, USA
| |
Collapse
|
12
|
Ramesh S, Cihalova D, Rajendran E, Van Dooren GG, Maier AG. Analysis of Plasmodium falciparum Mitochondrial Electron Transport Chain Activity Using Seahorse XFe96 Extracellular Flux Assays. Bio Protoc 2023; 13:e4863. [PMID: 37969754 PMCID: PMC10632155 DOI: 10.21769/bioprotoc.4863] [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: 05/02/2023] [Revised: 08/02/2023] [Accepted: 08/09/2023] [Indexed: 11/17/2023] Open
Abstract
The mitochondrial electron transport chain (ETC) is a multi-component pathway that mediates the transfer of electrons from metabolic reactions that occur in the mitochondrion to molecular oxygen (O2). The ETC contributes to numerous cellular processes, including the generation of cellular ATP through oxidative phosphorylation, serving as an electron sink for metabolic pathways such as de novo pyrimidine biosynthesis and for maintaining mitochondrial membrane potential. Proper functioning of the mitochondrial ETC is necessary for the growth and survival of apicomplexan parasites including Plasmodium falciparum, a causative agent of malaria. The mitochondrial ETC of P. falciparum is an attractive target for antimalarial drugs, due to its essentiality and its differences from the mammalian ETC. To identify novel P. falciparum ETC inhibitors, we have established a real-time assay to assess ETC function, which we describe here. This approach measures the O2 consumption rate (OCR) of permeabilized P. falciparum parasites using a Seahorse XFe96 flux analyzer and can be used to screen compound libraries for the identification of ETC inhibitors and, in part, to determine the targets of those inhibitors. Key features • With this protocol, the effects of candidate inhibitors on mitochondrial O2 consumption in permeabilized asexual P. falciparum parasites can be tested in real time. • Through the sequential injection of inhibitors and substrates into the assay, the molecular targets of candidate inhibitors in the ETC can, in part, be determined. • The assay is applicable for both drug discovery approaches and enquiries into a fundamental aspect of parasite mitochondrial biology.
Collapse
Affiliation(s)
- SaiShyam Ramesh
- Research School of Biology, The Australian National University, Canberra, Australia
| | - Daniela Cihalova
- Research School of Biology, The Australian National University, Canberra, Australia
| | - Esther Rajendran
- Research School of Biology, The Australian National University, Canberra, Australia
| | - Giel G. Van Dooren
- Research School of Biology, The Australian National University, Canberra, Australia
| | - Alexander G. Maier
- Research School of Biology, The Australian National University, Canberra, Australia
| |
Collapse
|
13
|
Klugherz I, Basch M, Ng N, Zhu Z, Wagener N, Wagener J. Only One of Three Bcs1 Homologs in Aspergillus fumigatus Confers Respiratory Growth. J Fungi (Basel) 2023; 9:1074. [PMID: 37998879 PMCID: PMC10672213 DOI: 10.3390/jof9111074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/25/2023] Open
Abstract
The mitochondrial translocase Bcs1 is required for the correct assembly of complex III of the mitochondrial respiratory chain. Because of its importance, Bcs1 was recently proposed as a target for antifungal agents. The function of this AAA (ATPase Associated with diverse cellular Activities) protein has been extensively characterized in Saccharomyces cerevisiae. This yeast as well as previously studied mammals each encode only one homolog. In contrast, the pathogenic mold Aspergillus fumigatus encodes three putative Bcs1 homologs, none of which have been characterized to date. To study the role of these three homologs in A. fumigatus, conditional and deletion mutants of the respective genes AFUA_3G13000 (bcs1A), AFUA_4G01260 (bcs1B), and AFUA_2G14760 (bcs1C) were generated. A deletion or downregulation of bcs1A resulted in drastically reduced growth and sporulation rates and in a significantly altered susceptibility to azole antifungals. In contrast, mutants lacking Bcs1B or Bcs1C did not show any phenotypes differing from the wild type. Salicylhydroxamic acid-an inhibitor of the alternative oxidase that allows the respiratory chain to bypass complex III in some species-caused a complete growth arrest of the bcs1A deletion mutant. In a Galleria mellonella infection model, the deletion of bcs1A resulted in significantly decreased virulence. Only Bcs1A was able to partially complement a deletion of BCS1 in S. cerevisiae. The subcellular localization of Bcs1B and Bcs1C outside of mitochondria suggests that these Bcs1 homologs exert cellular functions different from that of Bcs1. Our data demonstrate that Bcs1A is the sole Bcs1 ortholog in A. fumigatus.
Collapse
Affiliation(s)
- Isabel Klugherz
- Max von Pettenkofer-Institut für Hygiene und Medizinische Mikrobiologie, Medizinische Fakultät, Ludwig-Maximilians-Universität München, 80336 Munich, Germany; (I.K.)
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Marion Basch
- Zell- und Entwicklungsbiologie, Department Biologie II, Ludwig-Maximilians-Universität München, Planegg-Martinsried, 82152 Munich, Germany
| | - Natanya Ng
- Department of Clinical Microbiology, School of Medicine, Trinity College Dublin, The University of Dublin, St James’s Hospital Campus, D08 RX0X Dublin, Ireland
| | - Zhaojun Zhu
- Max von Pettenkofer-Institut für Hygiene und Medizinische Mikrobiologie, Medizinische Fakultät, Ludwig-Maximilians-Universität München, 80336 Munich, Germany; (I.K.)
| | - Nikola Wagener
- Zell- und Entwicklungsbiologie, Department Biologie II, Ludwig-Maximilians-Universität München, Planegg-Martinsried, 82152 Munich, Germany
| | - Johannes Wagener
- Max von Pettenkofer-Institut für Hygiene und Medizinische Mikrobiologie, Medizinische Fakultät, Ludwig-Maximilians-Universität München, 80336 Munich, Germany; (I.K.)
- Department of Clinical Microbiology, School of Medicine, Trinity College Dublin, The University of Dublin, St James’s Hospital Campus, D08 RX0X Dublin, Ireland
| |
Collapse
|
14
|
Rodrigues DS, Cabral VP, Barbosa AD, Valente Sá LG, Silva CR, Moreira LE, Neto JB, Silva J, Santos HS, Marinho ES, Cavalcanti BC, Moraes MO, Nobre Júnior HV. Sertraline has fungicidal activity against Candida spp. and acts by inhibiting membrane and cell wall biosynthesis. Future Microbiol 2023; 18:1025-1039. [PMID: 37540066 DOI: 10.2217/fmb-2022-0254] [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] [Indexed: 08/05/2023] Open
Abstract
Aim: Our study evaluated the activity of sertraline (SER) alone and associated with antifungal drugs in planktonic Candida spp. strains, and investigated its mechanism of action. Materials & methods: Broth microdilution method and minimum fungicidal concentration/MIC ratio were used to assess SER anticandidal activity, and the interaction with antifungals was determined by fractional inhibitory concentration index. The mechanism of action was investigated by flow cytometry and in silico tests. Results: SER inhibited Candida spp. strains at low concentrations by the fungicidal effect and showed no loss of effectiveness when combined. Its action seemed to be related to the membrane and cell wall biosynthesis inhibition. Conclusion: SER has activity against Candida spp. isolated and associated with antifungals, and acts by causing cell wall and membrane damage.
Collapse
Affiliation(s)
- Daniel S Rodrigues
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, 430-372, Brazil
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, 430-275, Brazil
| | - Vitória Pf Cabral
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, 430-372, Brazil
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, 430-275, Brazil
| | - Amanda D Barbosa
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, 430-372, Brazil
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, 430-275, Brazil
| | - Lívia Ga Valente Sá
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, 430-372, Brazil
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, 430-275, Brazil
- Christus University Center (UNICHRISTUS), Fortaleza, CE, Brazil
| | - Cecília R Silva
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, 430-372, Brazil
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, 430-275, Brazil
| | - Lara Ea Moreira
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, 430-372, Brazil
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, 430-275, Brazil
| | - Joao Ba Neto
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, 430-372, Brazil
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, 430-275, Brazil
- Christus University Center (UNICHRISTUS), Fortaleza, CE, Brazil
| | - Jacilene Silva
- Department of Chemistry, Group of Theoretical Chemistry and Electrochemistry (GQTE), State University of Ceará, Limoeiro do Norte, Ceará, 930-000, Brazil
| | - Hélcio S Santos
- Science and Technology Center, Chemistry Course, Vale do Acaraú State University, CE, 040-370, Sobral
| | - Emmanuel S Marinho
- Department of Chemistry, Group of Theoretical Chemistry and Electrochemistry (GQTE), State University of Ceará, Limoeiro do Norte, Ceará, 930-000, Brazil
| | - Bruno C Cavalcanti
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, 430-275, Brazil
| | - Manoel O Moraes
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, 430-275, Brazil
| | - Hélio V Nobre Júnior
- School of Pharmacy, Laboratory of Bioprospection in Antimicrobial Molecules (LABIMAN), Federal University of Ceará, Fortaleza, CE, 430-372, Brazil
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, 430-275, Brazil
| |
Collapse
|
15
|
Samartsev VN, Semenova AA, Belosludtsev KN, Dubinin MV. Modulators reducing the efficiency of oxidative ATP synthesis in mitochondria: protonophore uncouplers, cyclic redox agents, and decouplers. Biophys Rev 2023; 15:851-857. [PMID: 37974985 PMCID: PMC10643702 DOI: 10.1007/s12551-023-01160-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 09/25/2023] [Indexed: 11/19/2023] Open
Abstract
This work considers the main indicators of the oxidative phosphorylation efficiency in mitochondria: the ADP/O and H+/O ratios. Three groups of modulators that reduce the efficiency of oxidative phosphorylation are compared: protonophore uncouplers, cyclic redox compounds, and decouplers. It is noted that some of them are considered effective therapeutic agents. The paper analyzes the authors' original data on the mechanism of action of natural decouplers, represented by long-chain α,ω-dioic acids, as antioxidants. In conclusion, we discuss the hypothesis of their participation in the rescue of hepatocytes in various disorders of carbohydrate and lipid metabolism.
Collapse
Affiliation(s)
| | - Alena A. Semenova
- Mari State University, pl. Lenina 1, Yoshkar-Ola, Mari El 424001 Russia
| | - Konstantin N. Belosludtsev
- Mari State University, pl. Lenina 1, Yoshkar-Ola, Mari El 424001 Russia
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia
| | | |
Collapse
|
16
|
Yuan SSF, Wang YM, Chan LP, Hung AC, Nguyen HDH, Chen YK, Hu SCS, Lo S, Wang YY. IL-1RA promotes oral squamous cell carcinoma malignancy through mitochondrial metabolism-mediated EGFR/JNK/SOX2 pathway. J Transl Med 2023; 21:473. [PMID: 37461111 PMCID: PMC10351194 DOI: 10.1186/s12967-023-04343-9] [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: 05/30/2023] [Accepted: 07/10/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Interleukin-1 receptor antagonist (IL-1RA), a member of the IL-1 family, has diverse roles in cancer development. However, the role of IL-1RA in oral squamous cell carcinoma (OSCC), in particular the underlying mechanisms, remains to be elucidated. METHODS Tumor tissues from OSCC patients were assessed for protein expression by immunohistochemistry. Patient survival was evaluated by Kaplan-Meier curve analysis. Impact of differential IL-1RA expression on cultured OSCC cell lines was assessed in vitro by clonogenic survival, tumorsphere formation, soft agar colony formation, and transwell cell migration and invasion assays. Oxygen consumption rate was measured by Seahorse analyzer or multi-mode plate reader. PCR array was applied to screen human cancer stem cell-related genes, proteome array for phosphorylation status of kinases, and Western blot for protein expression in cultured cells. In vivo tumor growth was investigated by orthotopic xenograft in mice, and protein expression in xenograft tumors assessed by immunohistochemistry. RESULTS Clinical analysis revealed that elevated IL-1RA expression in OSCC tumor tissues was associated with increased tumor size and cancer stage, and reduced survival in the patient group receiving adjuvant radiotherapy compared to the patient group without adjuvant radiotherapy. In vitro data supported these observations, showing that overexpression of IL-1RA increased OSCC cell growth, migration/invasion abilities, and resistance to ionizing radiation, whereas knockdown of IL-1RA had largely the opposite effects. Additionally, we identified that EGFR/JNK activation and SOX2 expression were modulated by differential IL-1RA expression downstream of mitochondrial metabolism, with application of mitochondrial complex inhibitors suppressing these pathways. Furthermore, in vivo data revealed that treatment with cisplatin or metformin-a mitochondrial complex inhibitor and conventional therapy for type 2 diabetes-reduced IL-1RA-associated xenograft tumor growth as well as EGFR/JNK activation and SOX2 expression. This inhibitory effect was further augmented by combination treatment with cisplatin and metformin. CONCLUSIONS The current study suggests that IL-1RA promoted OSCC malignancy through mitochondrial metabolism-mediated EGFR/JNK activation and SOX2 expression. Inhibition of this mitochondrial metabolic pathway may present a potential therapeutic strategy in OSCC.
Collapse
Affiliation(s)
- Shyng-Shiou F Yuan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
- Translational Research Center, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, 75 Bo-Ai Street, Hsinchu, 300, Taiwan
| | - Yun-Ming Wang
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, 75 Bo-Ai Street, Hsinchu, 300, Taiwan
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, No.100, Shih-Chuan 1St Road, Sanmin Dist., Kaohsiung, 80708, Taiwan
| | - Leong-Perng Chan
- Cohort Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Department of Otorhinolaryngology-Head and Neck Surgery, Kaohsiung Municipal Ta-Tung Hospital and Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Amos C Hung
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
| | - Hieu D H Nguyen
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, No.100, Shih-Chuan 1St Road, Sanmin Dist., Kaohsiung, 80708, Taiwan
| | - Yuk-Kwan Chen
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, No.100, Shih-Chuan 1St Road, Sanmin Dist., Kaohsiung, 80708, Taiwan
- Division of Oral Pathology & Maxillofacial Radiology, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Stephen Chu-Sung Hu
- Department of Dermatology, College of Medicine, Kaohsiung Medical University, Kaohsiung, 807, Taiwan
- Department of Dermatology, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan
| | - Steven Lo
- Canniesburn Regional Plastic Surgery and Burns Unit, Glasgow, G4 0SF, UK
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Yen-Yun Wang
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 807, Taiwan.
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, 807, Taiwan.
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, No.100, Shih-Chuan 1St Road, Sanmin Dist., Kaohsiung, 80708, Taiwan.
| |
Collapse
|
17
|
do Nascimento FB, Valente Sá LG, de Andrade Neto JB, Cabral VP, Rodrigues DS, Barbosa AD, Moreira LE, Oliveira LC, Silva A, Lima IS, Silva J, Marinho ES, Santos HS, Cavalcanti BC, Morais MO, Júnior HV, Silva CR. Antifungal activity of cisatracurium against fluconazole-resistant Candida isolates and its antibiofilm effects. Future Microbiol 2023; 18:649-660. [PMID: 37522164 DOI: 10.2217/fmb-2022-0224] [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] [Indexed: 08/01/2023] Open
Abstract
Aim: To evaluate the antifungal activity of cisatracurium against Candida spp. resistant to fluconazole strains in planktonic and biofilm forms, in addition to determining its mechanism of action. Materials & methods: Antifungal activity and pharmacological interactions were determined using broth microdilution methods and the mechanism of action was evaluated by flow cytometry and molecular docking. Results: Cisatracurium presented antifungal activity against Candida spp. planktonic cells due to alterations of mitochondrial transmembrane potential leading to cellular apoptosis in addition to interacting with important targets related to cellular respiration, membrane and cell wall evidenced by molecular docking. Furthermore, the drug both prevented biofilm formation and impaired mature biofilms. Conclusion: Cisatracurium exhibits potential antifungal activity against Candida spp.
Collapse
Affiliation(s)
- Francisca Ba do Nascimento
- School of Pharmacy, Federal University of Ceará, Fortaleza, CEP, 60.430-372, Brazil
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil, CE, CEP, 60.430-275, Brasil
| | - Lívia Ga Valente Sá
- School of Pharmacy, Federal University of Ceará, Fortaleza, CEP, 60.430-372, Brazil
- Christus University Center, Fortaleza, CE, Brasil
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil, CE, CEP, 60.430-275, Brasil
| | - João B de Andrade Neto
- School of Pharmacy, Federal University of Ceará, Fortaleza, CEP, 60.430-372, Brazil
- Christus University Center, Fortaleza, CE, Brasil
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil, CE, CEP, 60.430-275, Brasil
| | - Vitória Pf Cabral
- School of Pharmacy, Federal University of Ceará, Fortaleza, CEP, 60.430-372, Brazil
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil, CE, CEP, 60.430-275, Brasil
| | - Daniel S Rodrigues
- School of Pharmacy, Federal University of Ceará, Fortaleza, CEP, 60.430-372, Brazil
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil, CE, CEP, 60.430-275, Brasil
| | - Amanda D Barbosa
- School of Pharmacy, Federal University of Ceará, Fortaleza, CEP, 60.430-372, Brazil
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil, CE, CEP, 60.430-275, Brasil
| | - Lara Ea Moreira
- School of Pharmacy, Federal University of Ceará, Fortaleza, CEP, 60.430-372, Brazil
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil, CE, CEP, 60.430-275, Brasil
| | - Leilson C Oliveira
- School of Pharmacy, Federal University of Ceará, Fortaleza, CEP, 60.430-372, Brazil
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil, CE, CEP, 60.430-275, Brasil
| | - Anderson Silva
- Institute of Advanced Chemistry, Higher Council for Scientific Research, Spain
| | - Iri Sp Lima
- Faculty of Medicine, Federal University of Ceará, Barbalha, 63048-080, CE, Brasil
| | - Jacilene Silva
- Theoretical Chemistry & Electrochemistry Group, State University of Ceará, Limoeiro do Norte, Ceará, 62.930-000, Brasil
| | - Emmanuel S Marinho
- Theoretical Chemistry & Electrochemistry Group, State University of Ceará, Limoeiro do Norte, Ceará, 62.930-000, Brasil
| | - Hélcio S Santos
- Science and Technology Centre, Course of Chemistry, State University Vale do Acaraú, Sobral, CE, Brazil
| | - Bruno C Cavalcanti
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil, CE, CEP, 60.430-275, Brasil
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil.CEP, 60.430-275, Brasil
| | - Manoel O Morais
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil, CE, CEP, 60.430-275, Brasil
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, CE, Brazil.CEP, 60.430-275, Brasil
| | - Hélio Vn Júnior
- School of Pharmacy, Federal University of Ceará, Fortaleza, CEP, 60.430-372, Brazil
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil, CE, CEP, 60.430-275, Brasil
| | - Cecília R Silva
- School of Pharmacy, Federal University of Ceará, Fortaleza, CEP, 60.430-372, Brazil
- Drug Research and Development Center, Federal University of Ceará, Fortaleza, CE, Brazil, CE, CEP, 60.430-275, Brasil
| |
Collapse
|
18
|
Hayward JA, Makota FV, Cihalova D, Leonard RA, Rajendran E, Zwahlen SM, Shuttleworth L, Wiedemann U, Spry C, Saliba KJ, Maier AG, van Dooren GG. A screen of drug-like molecules identifies chemically diverse electron transport chain inhibitors in apicomplexan parasites. PLoS Pathog 2023; 19:e1011517. [PMID: 37471441 PMCID: PMC10403144 DOI: 10.1371/journal.ppat.1011517] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/04/2023] [Accepted: 06/28/2023] [Indexed: 07/22/2023] Open
Abstract
Apicomplexans are widespread parasites of humans and other animals, and include the causative agents of malaria (Plasmodium species) and toxoplasmosis (Toxoplasma gondii). Existing anti-apicomplexan therapies are beset with issues around drug resistance and toxicity, and new treatment options are needed. The mitochondrial electron transport chain (ETC) is one of the few processes that has been validated as a drug target in apicomplexans. To identify new inhibitors of the apicomplexan ETC, we developed a Seahorse XFe96 flux analyzer approach to screen the 400 compounds contained within the Medicines for Malaria Venture 'Pathogen Box' for ETC inhibition. We identified six chemically diverse, on-target inhibitors of the ETC in T. gondii, at least four of which also target the ETC of Plasmodium falciparum. Two of the identified compounds (MMV024937 and MMV688853) represent novel ETC inhibitor chemotypes. MMV688853 belongs to a compound class, the aminopyrazole carboxamides, that were shown previously to target a kinase with a key role in parasite invasion of host cells. Our data therefore reveal that MMV688853 has dual targets in apicomplexans. We further developed our approach to pinpoint the molecular targets of these inhibitors, demonstrating that all target Complex III of the ETC, with MMV688853 targeting the ubiquinone reduction (Qi) site of the complex. Most of the compounds we identified remain effective inhibitors of parasites that are resistant to Complex III inhibitors that are in clinical use or development, indicating that they could be used in treating drug resistant parasites. In sum, we have developed a versatile, scalable approach to screen for compounds that target the ETC in apicomplexan parasites, and used this to identify and characterize novel inhibitors.
Collapse
Affiliation(s)
- Jenni A. Hayward
- Research School of Biology, Australian National University, Canberra, Australia
| | - F. Victor Makota
- Research School of Biology, Australian National University, Canberra, Australia
| | - Daniela Cihalova
- Research School of Biology, Australian National University, Canberra, Australia
| | - Rachel A. Leonard
- Research School of Biology, Australian National University, Canberra, Australia
| | - Esther Rajendran
- Research School of Biology, Australian National University, Canberra, Australia
| | - Soraya M. Zwahlen
- Research School of Biology, Australian National University, Canberra, Australia
| | - Laura Shuttleworth
- Research School of Biology, Australian National University, Canberra, Australia
| | - Ursula Wiedemann
- Research School of Biology, Australian National University, Canberra, Australia
| | - Christina Spry
- Research School of Biology, Australian National University, Canberra, Australia
| | - Kevin J. Saliba
- Research School of Biology, Australian National University, Canberra, Australia
| | - Alexander G. Maier
- Research School of Biology, Australian National University, Canberra, Australia
| | - Giel G. van Dooren
- Research School of Biology, Australian National University, Canberra, Australia
| |
Collapse
|
19
|
Zhang J, Fernando SD. Identification of Fungicide Combinations Targeting Plasmopara viticola and Botrytis cinerea Fungicide Resistance Using Machine Learning. Microorganisms 2023; 11:1341. [PMID: 37317315 DOI: 10.3390/microorganisms11051341] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 06/16/2023] Open
Abstract
Downy mildew (caused by Plasmopara viticola) and gray mold (caused by Botrytis cinerea) are fungal diseases that significantly impact grape production globally. Cytochrome b plays a significant role in the mitochondrial respiratory chain of the two fungi that cause these diseases and is a key target for quinone outside inhibitor (QoI)-based fungicide development. Since the mode of action (MOA) of QoI fungicides is restricted to a single active site, the risk of developing resistance to these fungicides is deemed high. Consequently, using a combination of fungicides is considered an effective way to reduce the development of QoI resistance. Currently, there is little information available to help in the selection of appropriate fungicides. This study used a combination of in silico simulations and quantitative structure-activity relationship (QSAR) machine learning algorithms to screen the most potent QoI-based fungicide combinations for wild-type (WT) and the G143A mutation of fungal cytochrome b. Based on in silico studies, mandestrobin emerged as the top binder for both WT Plasmopara viticola and WT Botrytis cinerea cytochrome b. Famoxadone appeared to be a versatile binder for G143A-mutated cytochrome b of both Plasmopara viticola and Botrytis cinerea. Thiram emerged as a reasonable, low-risk non-QoI fungicide that works on WT and G143A-mutated versions of both fungi. QSAR analysis revealed fenpropidin, fenoxanil, and ethaboxam non-QoIs to have a high affinity for G143A-mutated cytochrome b of Plasmopara viticola and Botrytis cinerea. Above-QoI and non-QoI fungicides can be considered for field studies in a fungicide management program against Plasmopara viticola- and Botrytis cinerea-based fungal infections.
Collapse
Affiliation(s)
- Junrui Zhang
- Biological and Agricultural Engineering Department, Texas A&M University, College Station, TX 77843-2117, USA
| | - Sandun D Fernando
- Biological and Agricultural Engineering Department, Texas A&M University, College Station, TX 77843-2117, USA
| |
Collapse
|
20
|
Sertse D, You FM, Klymiuk V, Haile JK, N'Diaye A, Pozniak CJ, Cloutier S, Kagale S. Historical Selection, Adaptation Signatures, and Ambiguity of Introgressions in Wheat. Int J Mol Sci 2023; 24:ijms24098390. [PMID: 37176097 PMCID: PMC10179502 DOI: 10.3390/ijms24098390] [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: 04/08/2023] [Revised: 05/03/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Wheat was one of the crops domesticated in the Fertile Crescent region approximately 10,000 years ago. Despite undergoing recent polyploidization, hull-to-free-thresh transition events, and domestication bottlenecks, wheat is now grown in over 130 countries and accounts for a quarter of the world's cereal production. The main reason for its widespread success is its broad genetic diversity that allows it to thrive in different environments. To trace historical selection and hybridization signatures, genome scans were performed on two datasets: approximately 113K SNPs from 921 predominantly bread wheat accessions and approximately 110K SNPs from about 400 wheat accessions representing all ploidy levels. To identify environmental factors associated with the loci, a genome-environment association (GEA) was also performed. The genome scans on both datasets identified a highly differentiated region on chromosome 4A where accessions in the first dataset were dichotomized into a group (n = 691), comprising nearly all cultivars, wild emmer, and most landraces, and a second group (n = 230), dominated by landraces and spelt accessions. The grouping of cultivars is likely linked to their potential ancestor, bread wheat cv. Norin-10. The 4A region harbored important genes involved in adaptations to environmental conditions. The GEA detected loci associated with latitude and temperature. The genetic signatures detected in this study provide insight into the historical selection and hybridization events in the wheat genome that shaped its current genetic structure and facilitated its success in a wide spectrum of environmental conditions. The genome scans and GEA approaches applied in this study can help in screening the germplasm housed in gene banks for breeding, and for conservation purposes.
Collapse
Affiliation(s)
- Demissew Sertse
- Aquatic and Crop Resource Development, National Research Council Canada, Saskatoon, SK S7N 0W9, Canada
| | - Frank M You
- Ottawa Research and Development Centre, Agriculture and Agri-Food Canada, Ottawa, ON K1A 0C6, Canada
| | - Valentyna Klymiuk
- Crop Development Centre, University of Saskatchewan, Saskatoon, SK S7N 5A8, Canada
| | - Jemanesh K Haile
- Crop Development Centre, University of Saskatchewan, Saskatoon, SK S7N 5A8, Canada
| | - Amidou N'Diaye
- Crop Development Centre, University of Saskatchewan, Saskatoon, SK S7N 5A8, Canada
| | - Curtis J Pozniak
- Crop Development Centre, University of Saskatchewan, Saskatoon, SK S7N 5A8, Canada
| | - Sylvie Cloutier
- Ottawa Research and Development Centre, Agriculture and Agri-Food Canada, Ottawa, ON K1A 0C6, Canada
| | - Sateesh Kagale
- Aquatic and Crop Resource Development, National Research Council Canada, Saskatoon, SK S7N 0W9, Canada
| |
Collapse
|
21
|
Zhan J, Xia D. Bcs1, a novel target for fungicide. Front Chem 2023; 11:1146753. [PMID: 36993815 PMCID: PMC10040684 DOI: 10.3389/fchem.2023.1146753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/03/2023] [Indexed: 03/14/2023] Open
Abstract
The mitochondrial respiratory chain has long been a primary target for the development of fungicides for its indispensable role in various cellular functions including energy metabolism. Over the years, a wide range of natural and synthetic fungicides and pesticides targeting the respiratory chain complexes have been discovered or developed and used in agriculture and in medicine, which brought considerable economic gains but was also accompanied by the emergence of resistance to these compounds. To delay and overcome the onset of resistance, novel targets for fungicides development are actively being pursued. Mitochondrial AAA protein Bcs1 is necessary for the biogenesis of respiratory chain Complex III, also known as cyt bc1 complex, by delivering the last essential iron-sulfur protein subunit in its folded form to the cyt bc1 precomplex. Although no report on the phenotypes of knock-out Bcs1 has been reported in animals, pathogenic Bcs1 mutations cause Complex III deficiency and respiratory growth defects, which makes it a promising new target for the development of fungicides. Recent Cryo-EM and X-ray structures of mouse and yeast Bcs1 revealed the basic oligomeric states of Bcs1, shed light on the translocation mechanism of its substrate ISP, and provided the basis for structure-based drug design. This review summarizes the recent progress made on understanding the structure and function of Bcs1, proposes the use of Bcs1 as an antifungal target, and provides novel prospects for fungicides design by targeting Bcs1.
Collapse
|
22
|
Wang S, Wang Z, Wang X, Qu J, Li F, Ji C, Wu H. Histopathological and transcriptomic analyses reveal the reproductive endocrine-disrupting effects of decabromodiphenyl ethane (DBDPE) in mussel Mytilus galloprovincialis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 862:160724. [PMID: 36493811 DOI: 10.1016/j.scitotenv.2022.160724] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/21/2022] [Accepted: 12/02/2022] [Indexed: 06/17/2023]
Abstract
The novel brominated flame retardant DBDPE has become a widespread environmental contaminant and could affect reproductive endocrine system in vertebrates. However, information about reproductive endocrine-disrupting effects of DBDPE on invertebrates is totally unknown. In this study, mussels Mytilus galloprovincialis were exposed to 1, 10, 50, 200 and 500 μg/L DBDPE for 30 days. Histopathological and transcriptomic analyses were performed to assess the reproductive endocrine-disrupting effects of DBDPE in mussels and the potential mechanisms. DBDPE promoted the gametogenesis in mussels of both sexes according to histological observation, gender-specific gene expression (VERL and VCL) and histological morphometric parameter analysis. Transcriptomic analysis demonstrated that DBDPE suppressed the genes related to cholesterol homeostasis and transport in both sexes via different LRPs- and ABCs-mediated pathways. DBDPE also disturbed nongenomic signaling pathway including signaling cascades (GPR157-IP3-Ca2+) in males and secondary messengers (cGMP) in females, and subsequently altered the expression levels of reproductive genes (VMO1, ZAN, Banf1 and Hook1). Additionally, dysregulation of energy metabolism in male mussels induced by DBDPE might interfere with the reproductive endocrine system. Overall, this is the first report that DBDPE evoked reproductive endocrine-disruptions in marine mussels. These findings will provide important references for ecological risk assessment of DBDPE pollution in marine environment.
Collapse
Affiliation(s)
- Shuang Wang
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai 264003, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China; College of Life Sciences, Yantai University, Yantai 264005, PR China
| | - Zhiyu Wang
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai 264003, PR China
| | - Xumin Wang
- College of Life Sciences, Yantai University, Yantai 264005, PR China
| | - Jiangyong Qu
- College of Life Sciences, Yantai University, Yantai 264005, PR China
| | - Fei Li
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai 264003, PR China; Center for Ocean Mega-Science, Chinese Academy of Sciences (CAS), Qingdao 266071, PR China
| | - Chenglong Ji
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai 264003, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, PR China; Center for Ocean Mega-Science, Chinese Academy of Sciences (CAS), Qingdao 266071, PR China.
| | - Huifeng Wu
- CAS Key Laboratory of Coastal Environmental Processes and Ecological Remediation, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences (CAS), Shandong Key Laboratory of Coastal Environmental Processes, YICCAS, Yantai 264003, PR China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, PR China; Center for Ocean Mega-Science, Chinese Academy of Sciences (CAS), Qingdao 266071, PR China
| |
Collapse
|
23
|
Cherrad S, Gillet B, Dellinger J, Bellaton L, Roux P, Hernandez C, Steva H, Perrier L, Vacher S, Hughes S. New insights from short and long reads sequencing to explore cytochrome b variants in Plasmopara viticola populations collected from vineyards and related to resistance to complex III inhibitors. PLoS One 2023; 18:e0268385. [PMID: 36656908 PMCID: PMC9851517 DOI: 10.1371/journal.pone.0268385] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 12/27/2022] [Indexed: 01/20/2023] Open
Abstract
Downy mildew is caused by Plasmopara viticola, an obligate oomycete plant pathogen, a devasting disease of grapevine. To protect plants from the disease, complex III inhibitors are among the fungicides widely used. They specifically target the mitochondrial cytochrome b (cytb) of the pathogen to block cellular respiration mechanisms. In the French vineyard, P. viticola has developed resistance against a first group of these fungicides, the Quinone outside Inhibitors (QoI), with a single amino acid substitution G143A in its cytb mitochondrial sequence. The use of QoI was limited and another type of fungicide, the Quinone inside Inhibitors, targeting the same gene and highly effective against oomycetes, was used instead. Recently however, less sensitive P. viticola populations were detected after treatments with some inhibitors, in particular ametoctradin and cyazofamid. By isolating single-sporangia P. viticola strains resistant to these fungicides, we characterized new variants in the cytb sequences associated with cyazofamid resistance: a point mutation (L201S) and more strikingly, two insertions (E203-DE-V204, E203-VE-V204). In parallel with the classical tools, pyrosequencing and qPCR, we then benchmarked short and long-reads NGS technologies (Ion Torrent, Illumina, Oxford Nanopore Technologies) to sequence the complete cytb with a view to detecting and assessing the proportion of resistant variants of P. viticola at the scale of a field population. Eighteen populations collected from French vineyard fields in 2020 were analysed: 12 showed a variable proportion of G143A, 11 of E203-DE-V204 and 7 populations of the S34L variant that confers resistance to ametoctradin. Interestingly, the long reads were able to identify variants, including SNPs, with confidence and to detect a small proportion of P. viticola with multiple variants along the same cytb sequence. Overall, NGS appears to be a promising method for assessing fungicide resistance of pathogens linked to cytb modifications at the field population level. This approach could rapidly become a robust decision support tool for resistance management in the future.
Collapse
Affiliation(s)
| | - Benjamin Gillet
- Institut de Génomique Fonctionnelle de Lyon (IGFL), CNRS UMR 5242, Ecole Normale Supérieure de Lyon, INRAE USC 1370, Université Claude Bernard Lyon 1, Lyon, France
| | - Julien Dellinger
- Institut de Génomique Fonctionnelle de Lyon (IGFL), CNRS UMR 5242, Ecole Normale Supérieure de Lyon, INRAE USC 1370, Université Claude Bernard Lyon 1, Lyon, France
| | - Lalie Bellaton
- Institut de Génomique Fonctionnelle de Lyon (IGFL), CNRS UMR 5242, Ecole Normale Supérieure de Lyon, INRAE USC 1370, Université Claude Bernard Lyon 1, Lyon, France
| | - Pascale Roux
- Institut de Génomique Fonctionnelle de Lyon (IGFL), CNRS UMR 5242, Ecole Normale Supérieure de Lyon, INRAE USC 1370, Université Claude Bernard Lyon 1, Lyon, France
| | | | | | | | | | - Sandrine Hughes
- Institut de Génomique Fonctionnelle de Lyon (IGFL), CNRS UMR 5242, Ecole Normale Supérieure de Lyon, INRAE USC 1370, Université Claude Bernard Lyon 1, Lyon, France
- * E-mail: (SH); (SC)
| |
Collapse
|
24
|
van der Watt ME, Reader J, Birkholtz LM. Adapt or Die: Targeting Unique Transmission-Stage Biology for Malaria Elimination. Front Cell Infect Microbiol 2022; 12:901971. [PMID: 35755845 PMCID: PMC9218253 DOI: 10.3389/fcimb.2022.901971] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/06/2022] [Indexed: 12/25/2022] Open
Abstract
Plasmodium parasites have a complex life cycle that includes development in the human host as well as the Anopheles vector. Successful transmission of the parasite between its host and vector therefore requires the parasite to balance its investments in asexual replication and sexual reproduction, varying the frequency of sexual commitment to persist within the human host and generate future opportunities for transmission. The transmission window is extended further by the ability of stage V gametocytes to circulate in peripheral blood for weeks, whereas immature stage I to IV gametocytes sequester in the bone marrow and spleen until final maturation. Due to the low gametocyte numbers in blood circulation and with the ease of targeting such life cycle bottlenecks, transmission represents an efficient target for therapeutic intervention. The biological process of Plasmodium transmission is a multistage, multifaceted process and the past decade has seen a much deeper understanding of the molecular mechanisms and regulators involved. Clearly, specific and divergent processes are used during transmission compared to asexual proliferation, which both poses challenges but also opportunities for discovery of transmission-blocking antimalarials. This review therefore presents an update of our molecular understanding of gametocyte and gamete biology as well as the status of transmission-blocking activities of current antimalarials and lead development compounds. By defining the biological components associated with transmission, considerations for the development of new transmission-blocking drugs to target such untapped but unique biology is suggested as an important, main driver for transmission-blocking drug discovery.
Collapse
Affiliation(s)
- Mariëtte E van der Watt
- Institute for Sustainable Malaria Control, School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa
| | - Janette Reader
- Institute for Sustainable Malaria Control, School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa.,Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
| | - Lyn-Marié Birkholtz
- Institute for Sustainable Malaria Control, School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa.,Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
25
|
Quinone binding sites of cyt bc complexes analysed by X-ray crystallography and cryogenic electron microscopy. Biochem Soc Trans 2022; 50:877-893. [PMID: 35356963 PMCID: PMC9162462 DOI: 10.1042/bst20190963] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/06/2022] [Accepted: 03/11/2022] [Indexed: 11/17/2022]
Abstract
Cytochrome (cyt) bc1, bcc and b6f complexes, collectively referred to as cyt bc complexes, are homologous isoprenoid quinol oxidising enzymes present in diverse phylogenetic lineages. Cyt bc1 and bcc complexes are constituents of the electron transport chain (ETC) of cellular respiration, and cyt b6f complex is a component of the photosynthetic ETC. Cyt bc complexes share in general the same Mitchellian Q cycle mechanism, with which they accomplish proton translocation and thus contribute to the generation of proton motive force which drives ATP synthesis. They therefore require a quinol oxidation (Qo) and a quinone reduction (Qi) site. Yet, cyt bc complexes evolved to adapt to specific electrochemical properties of different quinone species and exhibit structural diversity. This review summarises structural information on native quinones and quinone-like inhibitors bound in cyt bc complexes resolved by X-ray crystallography and cryo-EM structures. Although the Qi site architecture of cyt bc1 complex and cyt bcc complex differs considerably, quinone molecules were resolved at the respective Qi sites in very similar distance to haem bH. In contrast, more diverse positions of native quinone molecules were resolved at Qo sites, suggesting multiple quinone binding positions or captured snapshots of trajectories toward the catalytic site. A wide spectrum of inhibitors resolved at Qo or Qi site covers fungicides, antimalarial and antituberculosis medications and drug candidates. The impact of these structures for characterising the Q cycle mechanism, as well as their relevance for the development of medications and agrochemicals are discussed.
Collapse
|
26
|
Progress Report: Antimicrobial Drug Discovery in the Resistance Era. Pharmaceuticals (Basel) 2022; 15:ph15040413. [PMID: 35455410 PMCID: PMC9030565 DOI: 10.3390/ph15040413] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 02/05/2023] Open
Abstract
Antibiotic resistance continues to be a most serious threat to public health. This situation demands that the scientific community increase their efforts for the discovery of alternative strategies to circumvent the problems associated with conventional small molecule therapeutics. The Global Antimicrobial Resistance and Use Surveillance System (GLASS) Report (published in June 2021) discloses the rapidly increasing number of bacterial infections that are mainly caused by antimicrobial-resistant bacteria. These concerns have initiated various government agencies and other organizations to educate the public regarding the appropriate use of antibiotics. This review discusses a brief highlight on the timeline of antimicrobial drug discovery with a special emphasis on the historical development of antimicrobial resistance. In addition, new antimicrobial targets and approaches, recent developments in drug screening, design, and delivery were covered. This review also discusses the emergence and roles of various antibiotic adjuvants and combination therapies while shedding light on current challenges and future perspectives. Overall, the emergence of resistant microbial strains has challenged drug discovery but their efforts to develop alternative technologies such as nanomaterials seem to be promising for the future.
Collapse
|
27
|
Lin S, Liu C, Zhao X, Han X, Li X, Ye Y, Li Z. Recent Advances of Pyridinone in Medicinal Chemistry. Front Chem 2022; 10:869860. [PMID: 35402370 PMCID: PMC8984125 DOI: 10.3389/fchem.2022.869860] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 03/02/2022] [Indexed: 12/11/2022] Open
Abstract
Pyridinones have been adopted as an important block in medicinal chemistry that could serve as hydrogen bond donors and acceptors. With the help of feasible synthesis routes via established condensation reactions, the physicochemical properties of such a scaffold could be manipulated by adjustment of polarity, lipophilicity, and hydrogen bonding, and eventually lead to its wide application in fragment-based drug design, biomolecular mimetics, and kinase hinge-binding motifs. In addition, most pyridinone derivatives exhibit various biological activities ranging from antitumor, antimicrobial, anti-inflammatory, and anticoagulant to cardiotonic effects. This review focuses on recent contributions of pyridinone cores to medicinal chemistry, and addresses the structural features and structure–activity relationships (SARs) of each drug-like molecule. These advancements contribute to an in-depth understanding of the potential of this biologically enriched scaffold and expedite the development of its new applications in drug discovery.
Collapse
Affiliation(s)
- Shibo Lin
- Department of Pharmacy, Chengdu Second People’s Hospital, Chengdu, China
- *Correspondence: Shibo Lin,
| | - Chun Liu
- Department of Pharmacy, Chengdu Second People’s Hospital, Chengdu, China
| | - Xiaotian Zhao
- Department of Pharmacy, Chengdu Second People’s Hospital, Chengdu, China
| | - Xiao Han
- Department of Pharmacy, Chengdu Second People’s Hospital, Chengdu, China
| | - Xuanhao Li
- Department of Pharmacy, Chengdu Second People’s Hospital, Chengdu, China
| | - Yongqin Ye
- Department of Pharmacy, Chengdu Second People’s Hospital, Chengdu, China
| | - Zheyu Li
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, School of Pharmacy, Chengdu University, Chengdu, China
| |
Collapse
|
28
|
Li BY, Kang GQ, Huang M, Duan WG, Lin GS, Huang M, Wang X. Synthesis, bioactivity and computational simulation study of novel (Z)-3-caren-5-one oxime ethers as potential antifungal agents. RESEARCH ON CHEMICAL INTERMEDIATES 2022. [DOI: 10.1007/s11164-022-04690-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
29
|
Song X, Jiang Z, Li J, Lu X, Han Q, Zhu K, Li H, Ling Y, Duan H. Synthesis, antifungal activity, and molecular dynamics study of novel geranyl aromatic sulfonamide compounds as potential complex III inhibitors. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02864-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
30
|
Cowger C, Meyers E, Whetten R. Sensitivity of the U.S. Wheat Powdery Mildew Population to Quinone Outside Inhibitor Fungicides and Determination of the Complete Blumeria graminis f. sp. tritici Cytochrome b Gene. PHYTOPATHOLOGY 2022; 112:249-260. [PMID: 34156265 DOI: 10.1094/phyto-04-21-0132-r] [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: 06/13/2023]
Abstract
Wheat powdery mildew, caused by Blumeria graminis f. sp. tritici, is managed primarily with cultivar resistance and foliar fungicides. Quinone outside inhibitors (QoIs), which target the mitochondrial cytochrome b (cytb) gene, are one of the two main fungicide classes used on wheat. While European populations of B. graminis f. sp. tritici are widely insensitive to QoIs, largely because of the cytb mutation G143A, the QoI sensitivity of the U.S. B. graminis f. sp. tritici population had never been evaluated despite years of QoI use on U.S. wheat. A total of 381 B. graminis f. sp. tritici isolates from 15 central and eastern U.S. states were screened for sensitivity to QoI fungicides pyraclostrobin and picoxystrobin. A modest range of sensitivities was observed, with maximum resistance factors of 11.2 for pyraclostrobin and 5.3 for picoxystrobin. The F129L, G137R, and G143A cytb mutations were not detected in the U.S. B. graminis f. sp. tritici population, nor were mutations identified in the PEWY loop, a key part of the Qo site. Thus, no genetic basis for the observed quantitative variation in QoI sensitivity of U.S. B. graminis f. sp. tritici was identified. Isolate sporulation was weakly negatively associated with reduced QoI sensitivity, suggesting a fitness cost. In the course of the study, the complete B. graminis f. sp. tritici cytb gene sequence was determined for the first time in the isolate 96224 v. 3.16 reference genome. Contrary to previous reports, the gene has an intron that appears to belong to intron group II, which is unusual in fungi. The study was the first QoI sensitivity screening of a large, geographically diverse set of U.S. B. graminis f. sp. tritici isolates, and while the population as a whole remains relatively sensitive, some quantitative loss of efficacy was observed.
Collapse
Affiliation(s)
- Christina Cowger
- Department of Entomology and Plant Pathology, North Carolina State University, Raleigh, NC 27695
- Agricultural Research Service, U.S. Department of Agriculture, Raleigh, NC 27695
| | - Emily Meyers
- Department of Entomology and Plant Pathology, North Carolina State University, Raleigh, NC 27695
| | - Rebecca Whetten
- Agricultural Research Service, U.S. Department of Agriculture, Raleigh, NC 27695
| |
Collapse
|
31
|
Yao C, Meyer KG, Gallup C, Bowling AJ, Hufnagl A, Myung K, Lutz J, Slanec T, Pence HE, Delgado J, Wang NX. Florylpicoxamid, a new picolinamide fungicide with broad spectrum activity. PEST MANAGEMENT SCIENCE 2021; 77:4483-4496. [PMID: 34010509 DOI: 10.1002/ps.6483] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 04/15/2021] [Accepted: 05/19/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Following the introduction of fenpicoxamid, a natural product-based fungicide targeting the Qi site of mitochondrial cytochrome bc1 complex, a second generation fully synthetic picolinamide, florylpicoxamid, was discovered and its biological activity and attributes were characterized. RESULTS In vitro fungal growth inhibition assays and in planta glasshouse biological activity evaluations showed florylpicoxamid was active against 21 different plant pathogenic fungi within the phyla Ascomycota and Basidiomycota. Among the pathogens evaluated, florylpicoxamid was most potent against Zymoseptoria tritici, the causal organism of wheat leaf blotch, providing 80% growth inhibition in vitro at 0.0046 mg L-1 and 80% disease control in planta at 0.03 mg L-1 when applied as a preventative treatment. Florylpicoxamid was more efficacious than epoxiconazole, fluxapyroxad, and benzovindiflupyr versus a Z. tritici wild-type isolate when applied as curative and preventative treatments, with superior 10-day curative reachback activity. Analytical studies and in planta tests demonstrated that florylpicoxamid partitioned into plants quickly and showed good systemicity and translaminar activity on both monocot and dicot plants. No cross-resistance was observed between florylpicoxamid and strobilurin or azole fungicides. Florylpicoxamid exerts its preventative effect by preventing spore germination on the leaf surface and curative activity by arresting mycelial growth and pycnidia development in leaf tissue. CONCLUSIONS With strong broad spectrum fungicidal activity, florylpicoxamid delivers an innovative solution for growers to sustain high productivity and quality of many crops, and also provides a new option for developing effective strategies for fungicide resistance management. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Chenglin Yao
- Corteva Agriscience, Crop Protection Discovery & Development, Indianapolis, IN, USA
| | - Kevin G Meyer
- Corteva Agriscience, Crop Protection Discovery & Development, Indianapolis, IN, USA
| | - Courtney Gallup
- Corteva Agriscience, Crop Protection Discovery & Development, Indianapolis, IN, USA
| | - Andrew J Bowling
- Corteva Agriscience, Crop Protection Discovery & Development, Indianapolis, IN, USA
| | - Andrea Hufnagl
- Corteva Agriscience, Crop Protection Discovery & Development, Guyancourt, France
| | | | - Jamie Lutz
- Corteva Agriscience, Crop Protection Discovery & Development, Indianapolis, IN, USA
| | - Thomas Slanec
- Corteva Agriscience, Crop Protection Discovery & Development, Indianapolis, IN, USA
| | - Heather E Pence
- Corteva Agriscience, Crop Protection Discovery & Development, Indianapolis, IN, USA
| | - Javier Delgado
- Corteva Agriscience, Crop Protection Discovery & Development, Indianapolis, IN, USA
| | - Nick X Wang
- Corteva Agriscience, Crop Protection Discovery & Development, Indianapolis, IN, USA
| |
Collapse
|
32
|
Fouché G, Michel T, Lalève A, Wang NX, Young DH, Meunier B, Debieu D, Fillinger S, Walker AS. Directed evolution predicts cytochrome b G37V target site modification as probable adaptive mechanism towards the QiI fungicide fenpicoxamid in Zymoseptoria tritici. Environ Microbiol 2021; 24:1117-1132. [PMID: 34490974 DOI: 10.1111/1462-2920.15760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/03/2021] [Accepted: 09/04/2021] [Indexed: 02/06/2023]
Abstract
Acquired resistance is a threat to antifungal efficacy in medicine and agriculture. The diversity of possible resistance mechanisms and highly adaptive traits of pathogens make it difficult to predict evolutionary outcomes of treatments. We used directed evolution as an approach to assess the resistance risk to the new fungicide fenpicoxamid in the wheat pathogenic fungus Zymoseptoria tritici. Fenpicoxamid inhibits complex III of the respiratory chain at the ubiquinone reduction site (Qi site) of the mitochondrially encoded cytochrome b, a different site than the widely used strobilurins which inhibit the same complex at the ubiquinol oxidation site (Qo site). We identified the G37V change within the cytochrome b Qi site as the most likely resistance mechanism to be selected in Z. tritici. This change triggered high fenpicoxamid resistance and halved the enzymatic activity of cytochrome b, despite no significant penalty for in vitro growth. We identified negative cross-resistance between isolates harbouring G37V or G143A, a Qo site change previously selected by strobilurins. Double mutants were less resistant to both QiIs and quinone outside inhibitors compared to single mutants. This work is a proof of concept that experimental evolution can be used to predict adaptation to fungicides and provides new perspectives for the management of QiIs.
Collapse
Affiliation(s)
- Guillaume Fouché
- Université Paris-Saclay, INRAE, AgroParisTech, UMR BIOGER, Thiverval-Grignon, 78850, France.,Corteva Agriscience, 9330 Zionsville Road, Indianapolis, IN, 46268, USA
| | - Thomas Michel
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, 91198, France
| | - Anaïs Lalève
- Université Paris-Saclay, INRAE, AgroParisTech, UMR BIOGER, Thiverval-Grignon, 78850, France
| | - Nick X Wang
- Corteva Agriscience, 9330 Zionsville Road, Indianapolis, IN, 46268, USA
| | - David H Young
- Corteva Agriscience, 9330 Zionsville Road, Indianapolis, IN, 46268, USA
| | - Brigitte Meunier
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, 91198, France
| | - Danièle Debieu
- Université Paris-Saclay, INRAE, AgroParisTech, UMR BIOGER, Thiverval-Grignon, 78850, France
| | - Sabine Fillinger
- Université Paris-Saclay, INRAE, AgroParisTech, UMR BIOGER, Thiverval-Grignon, 78850, France
| | - Anne-Sophie Walker
- Université Paris-Saclay, INRAE, AgroParisTech, UMR BIOGER, Thiverval-Grignon, 78850, France
| |
Collapse
|
33
|
Hawkins NJ, Fraaije BA. Contrasting levels of genetic predictability in the evolution of resistance to major classes of fungicides. Mol Ecol 2021; 30:5318-5327. [PMID: 33706414 DOI: 10.1111/mec.15877] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 03/05/2021] [Indexed: 12/14/2022]
Abstract
The evolution of resistance has been seen across all major classes of xenobiotics, including antimicrobial drugs and agricultural pesticides. This repeated emergence of resistance is a case of phenotypic parallel evolution, but often the parallelism extends to the molecular level too, with multiple species gaining the same mutation in response to the same chemical treatment. We review the degree of repeatability in target-site resistance mutations affecting different classes of site-specific agricultural fungicides used in crop protection, comparing the extent to which resistance in different pathogen species has evolved via the same or different mutations. For all major fungicide target sites, substantial levels of molecular parallel evolution can be seen, with at least one mutation recurring in over 50% of species. Target-site mutations appear to be most repeatable in cytochrome b, target site of quinone-outside inhibitor fungicides, and least predictable for CYP51, target site of the azoles. Intermediate levels of repeatability are seen for the MBC target site β-tubulin, and the SDHI target site succinate dehydrogenase. Repeatability may be lower where there are selective trade-offs between resistance and pleiotropic fitness penalties, or differing levels of cross-resistance across members of a fungicide class; or where single mutations confer only partial resistance, and epistatic interactions between multiple mutations result in a rugged fitness landscape. This affects the predictive power of in vitro mutation studies, and has practical implications for resistance monitoring strategies and diagnostic methods.
Collapse
Affiliation(s)
- Nichola J Hawkins
- Biointeractions and Crop Protection Department, Rothamsted Research, Harpenden, Hertfordshire, UK.,NIAB, Cambridge, UK
| | | |
Collapse
|
34
|
Natural Compound-derived Cytochrome bc1 Complex Inhibitors as Antifungal Agents. Molecules 2020; 25:molecules25194582. [PMID: 33036496 PMCID: PMC7583968 DOI: 10.3390/molecules25194582] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/25/2020] [Accepted: 10/06/2020] [Indexed: 01/09/2023] Open
Abstract
The high incidence of fungal pathogens has become a global issue for crop protection. A promising strategy to control fungal plant infections is based on the use of nature-inspired compounds. The cytochrome bc1 complex is an essential component of the cellular respiratory chain and is one of the most important fungicidal targets. Natural products have played a crucial role in the discovery of cytochrome bc1 inhibitors, as proven by the development of strobilurins, one of the most important classes of crop-protection agents, over the past two decades. In this review, we summarize advances in the exploration of natural product scaffolds for the design and development of new bc1 complex inhibitors. Particular emphasis is given to molecular modeling-based approaches and structure-activity relationship (SAR) studies performed to improve the stability and increase the potency of natural precursors. The collected results highlight the versatility of natural compounds and provide an insight into the potential development of nature-inspired derivatives as antifungal agents.
Collapse
|