1
|
Yaeger LN, Sychantha D, Luong P, Shekarriz S, Goncalves O, Dobrin A, Ranieri MR, Lamers RP, Harvey H, diCenzo GC, Surette M, Côté JP, Magolan J, Burrows LL. Perturbation of Pseudomonas aeruginosa peptidoglycan recycling by anti-folates and design of a dual-action inhibitor. mBio 2025; 16:e0298424. [PMID: 39878530 PMCID: PMC11898565 DOI: 10.1128/mbio.02984-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 12/17/2024] [Indexed: 01/31/2025] Open
Abstract
Peptidoglycan (PG) is an important bacterial macromolecule that confers cell shape and structural integrity, and is a key antibiotic target. Its synthesis and turnover are carefully coordinated with other cellular processes and pathways. Despite established connections between the biosynthesis of PG and the outer membrane, or PG and DNA replication, links between PG and folate metabolism remain comparatively unexplored. Folate is an essential cofactor for bacterial growth and is required for the synthesis of many important metabolites. Here we show that inhibition of folate synthesis in the important Gram-negative pathogen Pseudomonas aeruginosa has downstream effects on PG metabolism and integrity that can manifest as the formation of a subpopulation of round cells that can undergo explosive lysis. Folate inhibitors potentiated β-lactams by perturbation of PG recycling, reducing expression of the AmpC β-lactamase. Supporting this mechanism, folate inhibitors also synergized with fosfomycin, an inhibitor of MurA, the first committed step in PG synthesis that can be bypassed by PG recycling. These insights led to the design of a dual-active inhibitor that overcomes NDM-1 metallo-β lactamase-mediated meropenem resistance and synergizes with the folate inhibitor, trimethoprim. We show that folate and PG metabolism are intimately connected, and targeting this connection can overcome antibiotic resistance in Gram-negative pathogens. IMPORTANCE To combat the alarming global increase in superbugs amid the simultaneous scarcity of new drugs, we can create synergistic combinations of currently available antibiotics or chimeric molecules with dual activities, to minimize resistance. Here we show that older anti-folate drugs synergize with specific cell wall biosynthesis inhibitors to kill the priority pathogen, Pseudomonas aeruginosa. Anti-folate drugs caused a dose-dependent loss of rod cell shape followed by explosive lysis, and synergized with β-lactams that target D,D-carboxypeptidases required to tailor the cell wall. Anti-folates impaired cell wall recycling and subsequent downstream expression of the chromosomally encoded β-lactamase, AmpC, which normally destroys β-lactam antibiotics. Building on the anti-folate-like scaffold of a metallo-β-lactamase inhibitor, we created a new molecule, MLLB-2201, that potentiates β-lactams and anti-folates and restores meropenem activity against metallo-β-lactamase-expressing Escherichia coli. These strategies are useful ways to tackle the ongoing rise in dangerous bacterial pathogens.
Collapse
Affiliation(s)
- Luke N. Yaeger
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - David Sychantha
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Princeton Luong
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Shahrokh Shekarriz
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Océane Goncalves
- Département de Biologie, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Annamaria Dobrin
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Michael R. Ranieri
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Ryan P. Lamers
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Hanjeong Harvey
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - George C. diCenzo
- Department of Biology, Queen’s University, Kingston, Ontario, Canada
| | - Michael Surette
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Jean-Phiippe Côté
- Département de Biologie, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Jakob Magolan
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Lori L. Burrows
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
2
|
Bhambid M, Walunj SB, Anupama CA, Jain S, Mehta D, Arya A, Wagstaff KM, Panda A, Jans DA, Mohmmed A, Patankar S. Importin α inhibitors act against the differentiated stages of apicomplexan parasites Plasmodium falciparum and Toxoplasma gondii. J Antimicrob Chemother 2025; 80:485-495. [PMID: 39691987 DOI: 10.1093/jac/dkae434] [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: 07/15/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Nuclear import, dependent on the transporter importin α (IMPα), is a drug target for apicomplexan parasites Plasmodium falciparum and Toxoplasma gondii. Indeed, a panel of small molecule inhibit interactions between IMPα and nuclear localization signals (NLSs) in vitro and the growth of rapidly dividing stages (P. falciparum blood stages and T. gondii tachyzoites) in culture. OBJECTIVES As new drugs targeting multiple life cycle stages of both parasites are required, the panel of IMPα inhibitors was tested for their ability to inhibit nuclear transport in the rapidly dividing stages and the maturation of differentiated stages (P. falciparum gametocytes and T. gondii bradyzoites). METHODS Using biophysical assays, Bay 11-7082, a Bay 11-7085 structural analogue, was tested for inhibition of IMPα:NLS interactions. The effect of the panel of inhibitors on the nuclear localization of reporter proteins was analysed in both parasites using transfections and microscopy. Also, using microscopy, the effect of inhibitors on differentiated stages of both parasites was tested. RESULTS Bay 11-7085 can inhibit nuclear transport in tachyzoites, while GW5074 and Caffeic Acid Phenethyl Ester (CAPE) can inhibit nuclear transport in the blood stages. Interestingly, CAPE can strongly inhibit gametocyte maturation, and Bay 11-7082 and Bay 11-7085 weakly inhibit bradyzoite differentiation. CONCLUSIONS As differentiation of gametocytes and bradyzoites is dependent on the activation of gene expression triggered by the nuclear translocation of transcription factors, our work provides a 'proof of concept' that targeting nuclear import is a viable strategy for the development of therapeutics against multiple stages of apicomplexan parasites, some of which are recalcitrant to existing drugs.
Collapse
Affiliation(s)
- Manasi Bhambid
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Sujata B Walunj
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
- IITB-Monash Research Academy, IIT Bombay, Mumbai, India
| | - C A Anupama
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Shilpi Jain
- International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Diksha Mehta
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
- IITB-Monash Research Academy, IIT Bombay, Mumbai, India
| | - Anjali Arya
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Kylie M Wagstaff
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Ashutosh Panda
- International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- Department of Microbiology, All India Institute of Medical Sciences, New Delhi, India
| | - David A Jans
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Asif Mohmmed
- International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Swati Patankar
- Department of Biosciences & Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
3
|
Schwalen F, Ghadi C, Ibazizene L, Khan SU, Sopkova-de Oliveira Santos J, Weiswald LB, Voisin-Chiret AS, Meryet-Figuiere M, Kieffer C. UBE2N: Hope on the Cancer Front, How to Inhibit This Promising Target Prospect? J Med Chem 2025; 68:915-928. [PMID: 39806871 DOI: 10.1021/acs.jmedchem.4c01517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
UBE2N protein belongs to the UE2s family and plays a crucial role in DNA repair, making it an exciting target for the development of innovative anticancer therapies. With the aim of discovering UBE2N inhibitors (UBE2Ni), this perspective seeks to review and provide elements to guide the design of new compounds. We propose a chemoinformatic structural analysis of the protein and its areas of interaction with its different partners. While covalent UBE2Ni are the most advanced molecules in their development, noncovalent inhibitors offer significant advantages that could overcome the limitations of covalent ones, particularly in terms of selectivity. Lastly, to obtain a drug candidate, early assessment of the druggability of compounds is essential in a hit to lead process. For existing UBE2Ni, a critical challenge lies in their pharmacokinetic properties and will obviously have to be considered as early as possible to hope for an application in human therapy.
Collapse
Affiliation(s)
- Florian Schwalen
- Université de Caen Normandie, CERMN UR4258, Normandie Univ, F-14000 Caen, France
- Pharmacie, CHU Caen Normandie, 14033 Caen, France
| | - Côme Ghadi
- Université de Caen Normandie, CERMN UR4258, Normandie Univ, F-14000 Caen, France
| | - Léonie Ibazizene
- Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Université de Caen Normandie, 14076 Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, 14076 Caen, France
| | - Shafi Ullah Khan
- Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Université de Caen Normandie, 14076 Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, 14076 Caen, France
| | | | - Louis-Bastien Weiswald
- Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Université de Caen Normandie, 14076 Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, 14076 Caen, France
| | | | - Matthieu Meryet-Figuiere
- Inserm U1086 ANTICIPE (Interdisciplinary Research Unit for Cancer Prevention and Treatment), Normandie Univ, Université de Caen Normandie, 14076 Caen, France
- Comprehensive Cancer Center François Baclesse, UNICANCER, 14076 Caen, France
| | - Charline Kieffer
- Université de Caen Normandie, CERMN UR4258, Normandie Univ, F-14000 Caen, France
| |
Collapse
|
4
|
Kasti A, Katsas K, Nikolaki MD, Triantafyllou K. The Role and the Regulation of NLRP3 Inflammasome in Irritable Bowel Syndrome: A Narrative Review. Microorganisms 2025; 13:171. [PMID: 39858939 PMCID: PMC11767632 DOI: 10.3390/microorganisms13010171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Irritable bowel syndrome (IBS) is a chronic disorder of the gastrointestinal tract. Its pathogenesis involves multiple factors, including visceral hypersensitivity and immune activation. NLRP3 inflammasome is part of the nucleotide-binding oligomerization domain-like receptor (NLR) family, a crucial component of the innate immune system. Preclinical studies have demonstrated that inhibiting NLRP3 reduces visceral sensitivity and IBS symptoms, like abdominal pain, and diarrhea, suggesting that targeting the NLRP3 might represent a novel therapeutic approach for IBS. This review aims to assess the NLRP3 inhibitors (tranilast, β-hydroxybutyrate, Chang-Kang-fang, paeoniflorin, coptisine, BAY 11-7082, and Bifidobacterium longum), highlighting the signaling pathways, and their potential role in IBS symptoms management was assessed. Although premature, knowledge of the action of synthetic small molecules, phytochemicals, organic compounds, and probiotics might make NLRP3 a new therapeutic target in the quiver of physicians' therapeutic choices for IBS symptoms management.
Collapse
Affiliation(s)
- Arezina Kasti
- Department of Nutrition and Dietetics, Attikon University General Hospital, 12462 Athens, Greece; (A.K.); (K.K.); (M.D.N.)
| | - Konstantinos Katsas
- Department of Nutrition and Dietetics, Attikon University General Hospital, 12462 Athens, Greece; (A.K.); (K.K.); (M.D.N.)
| | - Maroulla D. Nikolaki
- Department of Nutrition and Dietetics, Attikon University General Hospital, 12462 Athens, Greece; (A.K.); (K.K.); (M.D.N.)
| | - Konstantinos Triantafyllou
- Hepatogastroenterology Unit, 2nd Department of Internal Propaedeutic Medicine, Medical School, National and Kapodistrian University of Athens, Attikon University General Hospital, 12462 Athens, Greece
| |
Collapse
|
5
|
Dyson PJ, Banat IM, Quinn GA. War and peace: exploring microbial defence systems as a source of new antimicrobial therapies. Front Pharmacol 2025; 15:1504901. [PMID: 39840088 PMCID: PMC11747395 DOI: 10.3389/fphar.2024.1504901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/13/2024] [Indexed: 01/23/2025] Open
Abstract
The WHO has compiled a list of pathogens that urgently require new antibiotics in response to the rising reports of antibiotic resistance and a diminished supply of new antibiotics. At the top of this list is fluoroquinolone-resistant Salmonella typhi, fluoroquinolone-resistant Shigella spp. and vancomycin-resistant Enterococcus faecium. Although these problems have been covered in great detail by other contemporary reviews, there are still some fundamental gaps in the translation of current knowledge of the infectious process and the molecular ecology of antibiotic production into a sustainable protocol for the treatment of pathogenic diseases. Therefore, in this narrative review we briefly discuss newly approved antimicrobial drugs (since 2014) that could help to alleviate the burden of multiresistant pathogens listed on the WHO priority list. Being conscious that such treatments may eventually run the risk of future cycles of resistance, we also discuss how new understandings in the molecular ecology of antibiotic production and the disease process can be harnessed to create a more sustainable solution for the treatment of pathogenic diseases.
Collapse
Affiliation(s)
- Paul J. Dyson
- Medical School, Institute of Life Sciences, Swansea University, Swansea, United Kingdom
| | - Ibrahim M. Banat
- Centre for Molecular Biosciences, Ulster University, Coleraine, United Kingdom
| | - Gerry A. Quinn
- Centre for Molecular Biosciences, Ulster University, Coleraine, United Kingdom
| |
Collapse
|
6
|
Giroud C, Szommer T, Coxon C, Monteiro O, Grimes T, Zarganes-Tzitzikas T, Christott T, Bennett J, Buchan K, Brennan PE, Fedorov O. Covalent Inhibitors of S100A4 Block the Formation of a Pro-Metastasis Non-Muscle Myosin 2A Complex. J Med Chem 2024; 67:18943-18956. [PMID: 39425667 PMCID: PMC11571109 DOI: 10.1021/acs.jmedchem.4c01320] [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: 06/20/2024] [Revised: 10/04/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
The S100 protein family functions as protein-protein interaction adaptors regulated by Ca2+ binding. Formation of various S100 complexes plays a central role in cell functions, from calcium homeostasis to cell signaling, and is implicated in cell growth, migration, and tumorigenesis. We established a suite of biochemical and cellular assays for small molecule screening based on known S100 protein-protein interactions. From 25 human S100 proteins, we focused our attention on S100A4 because of its well-established role in cancer progression and metastasizes by interacting with nonmuscle myosin II (NMII). We identified several potent and selective inhibitors of this interaction and established the covalent nature of binding, confirmed by mass spectrometry and crystal structures. 5b showed on-target activity in cells and inhibition of cancer cell migration. The identified S100A4 inhibitors can serve as a basis for the discovery of new cancer drugs operating via a novel mode of action.
Collapse
Affiliation(s)
- Charline Giroud
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - Tamas Szommer
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - Carmen Coxon
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - Octovia Monteiro
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - Thomas Grimes
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
- Alzheimer’s
Research UK Oxford Drug Discovery Institute, NDM Research Building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - Tryfon Zarganes-Tzitzikas
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
- Alzheimer’s
Research UK Oxford Drug Discovery Institute, NDM Research Building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - Thomas Christott
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - James Bennett
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - Karly Buchan
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - Paul E. Brennan
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
| | - Oleg Fedorov
- Centre
for Medicines Discovery, Nuffield Department
of Medicine, NDM Research
building, Old Road Campus, Oxford OX3 7FZ, U.K.
| |
Collapse
|
7
|
Sychantha D, Chen X, Koteva K, Prehna G, Wright GD. Targeting bacterial nickel transport with aspergillomarasmine A suppresses virulence-associated Ni-dependent enzymes. Nat Commun 2024; 15:4036. [PMID: 38740750 DOI: 10.1038/s41467-024-48232-1] [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: 06/02/2023] [Accepted: 04/23/2024] [Indexed: 05/16/2024] Open
Abstract
Microbial Ni2+ homeostasis underpins the virulence of several clinical pathogens. Ni2+ is an essential cofactor in urease and [NiFe]-hydrogenases involved in colonization and persistence. Many microbes produce metallophores to sequester metals necessary for their metabolism and starve competing neighboring organisms. The fungal metallophore aspergillomarasmine A (AMA) shows narrow specificity for Zn2+, Ni2+, and Co2+. Here, we show that this specificity allows AMA to block the uptake of Ni2+ and attenuate bacterial Ni-dependent enzymes, offering a potential strategy for reducing virulence. Bacterial exposure to AMA perturbs H2 metabolism, ureolysis, struvite crystallization, and biofilm formation and shows efficacy in a Galleria mellonella animal infection model. The inhibition of Ni-dependent enzymes was aided by Zn2+, which complexes with AMA and competes with the native nickelophore for the uptake of Ni2+. Biochemical analyses demonstrated high-affinity binding of AMA-metal complexes to NikA, the periplasmic substrate-binding protein of the Ni2+ uptake system. Structural examination of NikA in complex with Ni-AMA revealed that the coordination geometry of Ni-AMA mimics the native ligand, Ni-(L-His)2, providing a structural basis for binding AMA-metal complexes. Structure-activity relationship studies of AMA identified regions of the molecule that improve NikA affinity and offer potential routes for further developing this compound as an anti-virulence agent.
Collapse
Affiliation(s)
- David Sychantha
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, ON, Canada
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
- Department of Chemistry, University of Waterloo, Waterloo, ON, Canada
| | - Xuefei Chen
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, ON, Canada
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Kalinka Koteva
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, ON, Canada
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Gerd Prehna
- Department of Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - Gerard D Wright
- David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, ON, Canada.
- M.G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada.
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
8
|
Xiao YC, Chen FE. The vinyl sulfone motif as a structural unit for novel drug design and discovery. Expert Opin Drug Discov 2024; 19:239-251. [PMID: 37978948 DOI: 10.1080/17460441.2023.2284201] [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: 09/01/2023] [Accepted: 11/13/2023] [Indexed: 11/19/2023]
Abstract
INTRODUCTION Vinyl sulfones are a special sulfur-containing structural unit that have attracted considerable attention, owing to their important role in serving as key structural motifs of various biologically active compounds as well as serving as versatile building blocks for organic transformations. The synthetic strategy of vinyl sulfone derivatives has been substantially upgraded over the past 30 years, and the wide application of this functional group in drug design and discovery has been promoted. AREA COVERED In this review, the authors review the application of vinyl sulfones in drug discovery and select optimized compounds which might have significant impact or potential inspiration for drug design. EXPERT OPINION Vinyl sulfones have been reported to target various macromolecular targets via non-covalent or covalent interactions, including multiple kinases, tubulin, cysteine protease, transcription factor, and so on. Thus, it has been significantly applied as a privileged scaffold in the design of anticancer, anti-infective, anti-inflammatory, and neuroprotective agents. However, much work remains to be done to improve the drug-like properties, such as chemical and metabolic stability, ADME, and toxicity. Besides, the chemical space of vinyl sulfones needs to be expanded, including but not limited to the design of constrained endocyclic and exocyclic vinyl sulfones.
Collapse
Affiliation(s)
- You-Cai Xiao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Ministry of Education and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Fen-Er Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Ministry of Education and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
- Engineering Center of Catalysis and Synthesis for Chiral Molecules, Department of Chemistry, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Yaeger LN, French S, Brown ED, Côté JP, Burrows LL. Central metabolism is a key player in E. coli biofilm stimulation by sub-MIC antibiotics. PLoS Genet 2023; 19:e1011013. [PMID: 37917668 PMCID: PMC10645362 DOI: 10.1371/journal.pgen.1011013] [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: 07/22/2023] [Revised: 11/14/2023] [Accepted: 10/10/2023] [Indexed: 11/04/2023] Open
Abstract
Exposure of Escherichia coli to sub-inhibitory antibiotics stimulates biofilm formation through poorly characterized mechanisms. Using a high-throughput Congo Red binding assay to report on biofilm matrix production, we screened ~4000 E. coli K12 deletion mutants for deficiencies in this biofilm stimulation response. We screened using three different antibiotics to identify core components of the biofilm stimulation response. Mutants lacking acnA, nuoE, or lpdA failed to respond to sub-MIC cefixime and novobiocin, implicating central metabolism and aerobic respiration in biofilm stimulation. These genes are members of the ArcA/B regulon-controlled by a respiration-sensitive two-component system. Mutants of arcA and arcB had a 'pre-activated' phenotype, where biofilm formation was already high relative to wild type in vehicle control conditions, and failed to increase further with the addition of sub-MIC cefixime. Using a tetrazolium dye and an in vivo NADH sensor, we showed spatial co-localization of increased metabolic activity with sub-lethal concentrations of the bactericidal antibiotics cefixime and novobiocin. Supporting a role for respiratory stress, the biofilm stimulation response to cefixime and novobiocin was inhibited when nitrate was provided as an alternative electron acceptor. Deletion of a gene encoding part of the machinery for respiring nitrate abolished its ameliorating effects, and nitrate respiration increased during growth with sub-MIC cefixime. Finally, in probing the generalizability of biofilm stimulation, we found that the stimulation response to translation inhibitors, unlike other antibiotic classes, was minimally affected by nitrate supplementation, suggesting that targeting the ribosome stimulates biofilm formation in distinct ways. By characterizing the biofilm stimulation response to sub-MIC antibiotics at a systems level, we identified multiple avenues for design of therapeutics that impair bacterial stress management.
Collapse
Affiliation(s)
- Luke N. Yaeger
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Shawn French
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Eric D. Brown
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - Jean Philippe Côté
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Département de Biologie, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Lori L. Burrows
- Department of Biochemistry and Biomedical Sciences, and the Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
10
|
Zheng X, Zhao D, Jin Y, Liu Y, Liu D. Role of the NLRP3 inflammasome in gynecological disease. Biomed Pharmacother 2023; 166:115393. [PMID: 37660654 DOI: 10.1016/j.biopha.2023.115393] [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: 07/04/2023] [Revised: 08/20/2023] [Accepted: 08/26/2023] [Indexed: 09/05/2023] Open
Abstract
The NLR family pyrin domain containing 3 (NLRP3) inflammasome is involved in the innate immune system and is a three-part macromolecular complex comprising the NLRP3 protein, apoptosis-associated speck-like protein containing a CARD (ASC) and the cysteine protease pro-caspase-1. When the NLRP3 inflammasome is activated, it can produce interleukin (IL)- 1β and IL-18 and eventually lead to inflammatory cell pyroptosis. Related studies have demonstrated that the NLRP3 inflammasome can induce an immune response and is related to the occurrence and development of gynecological diseases, such as endometriosis, polycystic ovary syndrome and breast cancer. NLRP3 inflammasome inhibitors are beneficial for maintaining cellular homeostasis and tissue health and have been found effective in targeting some gynecological diseases. However, excessive inhibitor concentrations have been found to cause adverse effects. Therefore, proper control of NLRP3 inflammasome activity is critical. This paper summarizes the structure and function of the NLRP3 inflammasome and highlights the therapeutic potential of targeting it in gynecological diseases, such as endometriosis, polycystic ovary syndrome and breast cancer The application of NLRP3 inflammasome inhibitors is also discussed.
Collapse
Affiliation(s)
- Xu Zheng
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Dan Zhao
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Ye Jin
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China.
| | - Yang Liu
- Acupuncture department,Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130117, Jilin, China.
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China.
| |
Collapse
|
11
|
Konaklieva MI, Plotkin BJ. Utilization of Existing Human Kinase Inhibitors as Scaffolds in the Development of New Antimicrobials. Antibiotics (Basel) 2023; 12:1418. [PMID: 37760715 PMCID: PMC10525673 DOI: 10.3390/antibiotics12091418] [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: 08/22/2023] [Revised: 09/03/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
The prevalence and continuing expansion of drug resistance, both in clinical and community settings represents a major challenge for current antimicrobial therapy. The different approaches for addressing this challenge include (1) identification of novel antibacterials by repurposing of existing drugs originally that historically target host proteins; and (2) effect target switching through modification of existing antimicrobials. The focus of this manuscript is on these drug discovery strategies, with utility for development of new antimicrobials with different modes of action.
Collapse
Affiliation(s)
| | - Balbina J. Plotkin
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL 60515, USA;
| |
Collapse
|
12
|
Liu Y, Lei H, Zhang W, Xing Q, Liu R, Wu S, Liu Z, Yan Q, Li W, Liu X, Hu Y. Pyroptosis in renal inflammation and fibrosis: current knowledge and clinical significance. Cell Death Dis 2023; 14:472. [PMID: 37500614 PMCID: PMC10374588 DOI: 10.1038/s41419-023-06005-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/29/2023]
Abstract
Pyroptosis is a novel inflammatory form of regulated cell death (RCD), characterized by cell swelling, membrane rupture, and pro-inflammatory effects. It is recognized as a potent inflammatory response required for maintaining organismal homeostasis. However, excessive and persistent pyroptosis contributes to severe inflammatory responses and accelerates the progression of numerous inflammation-related disorders. In pyroptosis, activated inflammasomes cleave gasdermins (GSDMs) and generate membrane holes, releasing interleukin (IL)-1β/18, ultimately causing pyroptotic cell death. Mechanistically, pyroptosis is categorized into caspase-1-mediated classical pyroptotic pathway and caspase-4/5/11-mediated non-classical pyroptotic pathway. Renal fibrosis is a kidney disease characterized by the loss of structural and functional units, the proliferation of fibroblasts and myofibroblasts, and extracellular matrix (ECM) accumulation, which leads to interstitial fibrosis of the kidney tubules. Histologically, renal fibrosis is the terminal stage of chronic inflammatory kidney disease. Although there is a multitude of newly discovered information regarding pyroptosis, the regulatory roles of pyroptosis involved in renal fibrosis still need to be fully comprehended, and how to improve clinical outcomes remains obscure. Hence, this review systematically summarizes the novel findings regarding the role of pyroptosis in the pathogenesis of renal fibrosis and discusses potential biomarkers and drugs for anti-fibrotic therapeutic strategies.
Collapse
Affiliation(s)
- Ya Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Haibo Lei
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Wenyou Zhang
- Department of Pharmacy, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Qichang Xing
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Renzhu Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Shiwei Wu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Zheng Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Qingzi Yan
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Wencan Li
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Xiang Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China.
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China.
| | - Yixiang Hu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China.
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China.
| |
Collapse
|