1
|
Natala SR, Habas A, Stocking EM, Orry A, Abagyan R, Makale MT, Wrasidlo W. Structure based design, synthesis and identification of novel covalent reversible dual TLR2/TLR9 small molecule antagonists. Bioorg Med Chem Lett 2025; 124:130259. [PMID: 40294721 DOI: 10.1016/j.bmcl.2025.130259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/13/2025] [Accepted: 04/26/2025] [Indexed: 04/30/2025]
Abstract
Inflammation is a key driver of the onset and progression of neurodegenerative diseases and cancer and can be caused by aggregated proteins, injured neurons or synapses, dysregulation of inflammatory control mechanisms, and other factors. Tolllike receptors (TLRs) are important mediators of inflammatory pathways, and their activation leads to pro-inflammatory cytokine release by immune cells in the periphery or in the central nervous system (CNS). TLR2 and TLR9 are implicated in the inflammatory pathogenesis of CNS degenerative diseases such as Parkinson's Disease (PD) and amyotrophic lateral sclerosis (ALS). They are also held to be important in the etiology of certain malignancies like inflammatory pancreatic ductal adenocarcinoma and glioblastoma. Inactivation of TLR2/9 in animal models of neurodegeneration has reduced pathological markers and diminished neuronal loss, while in animal models of cancer it has suppressed tumors. Therefore, TLR2 and TLR9 may be potential targets for the treatment of neurodegenerative disorders and cancers. We identified for the first time a key binding locus in TLR2/9 TIR domain which guided reversible covalent drug (RCD) design of a novel, first-in class series of dual TLR2/9 antagonists. Sub-micromolar antagonist concentrations potently inhibited TLR2 and TLR9 signaling induced by TLR2/9 specific agonists. Importantly, this series of antagonists did not discernably activate other TLRs and exhibited favorable in-vitro ADME and safety. The analogs described here may help realize effective TLR2/9 antagonism as a viable therapeutic strategy for inflammation driven CNS diseases and various malignancies with an inflammatory etiology.
Collapse
Affiliation(s)
| | - Agata Habas
- Eos Therapies, Inc., 10601 Tierrasanta Boulevard, Suite G. PMB 227, San Diego, CA 92124, United States
| | - Emily M Stocking
- Eos Therapies, Inc., 10601 Tierrasanta Boulevard, Suite G. PMB 227, San Diego, CA 92124, United States
| | - Andrew Orry
- MolSoft LLC., 11199 Sorrento Valley Rd # 209, San Diego, CA 92121, USA
| | - Ruben Abagyan
- University of California, San Diego, Skaggs School of Pharmacy & Pharmaceutical Science, La Jolla, CA 92093, USA
| | - Milan T Makale
- University of California, San Diego, Radiation Medicine and Applied Sciences, La Jolla, CA 92093, United States
| | - Wolfgang Wrasidlo
- University of California, San Diego, Radiation Medicine and Applied Sciences, La Jolla, CA 92093, United States.
| |
Collapse
|
2
|
Song X, Wang H, Gao Y, Zhang W, Lei X. Synthesis and biological evaluation of the Fluoro analog of Romidepsin with improved selectivity for class I histone deacetylases (HDACs). Bioorg Chem 2025; 159:108348. [PMID: 40090152 DOI: 10.1016/j.bioorg.2025.108348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/17/2025] [Accepted: 03/04/2025] [Indexed: 03/18/2025]
Abstract
Selective inhibition of Class I HDACs has emerged as a promising approach for cancer therapy. Building on our previous work with Largazole (a member of the natural depsipeptide family), we have applied a similar fluorination modification to Romidepsin and synthesized its fluoro analog (12) in 12 steps. This analog exhibits potent inhibitory activity against Class I HDACs but shows no inhibitory effect on HDAC6, confirming its selectivity as a Class I HDAC inhibitor (IC50 HDAC1 0.95 nM, HDAC2 0.86, HDAC 3 1.1 nM, HDAC8 4.2 nM, HDAC6 > 103 nM). Compared with Romidepsin, compound 12 demonstrates significant growth inhibition in two cancer cell lines (NCI-H1975 and HT29) while exhibiting markedly less growth inhibition in two normal cell lines (WRL-68 and HEK293). Further studies reveal that 12 is capable of blocking the cell cycle and inducing apoptosis, thereby exerting anticancer activity. Moreover, 12 possesses metabolic stability comparable to Romidepsin. In a mouse model, 12 demonstrates strong in vivo antitumor efficacy similar to that of Romidepsin, yet with significantly reduced toxicity. These findings support the potential of this fluoro analog as a highly selective Class I HDAC inhibitor and highlight its promise as a superior alternative to Romidepsin for further development.
Collapse
Affiliation(s)
- Xinluo Song
- School of Pharmacy, Fudan University; 826 Zhangheng Road, Pudong Zone, Shanghai 201203, China
| | - Hanqi Wang
- School of Pharmacy, Fudan University; 826 Zhangheng Road, Pudong Zone, Shanghai 201203, China
| | - Ya Gao
- Shanghai Forxine Pharmaceutical Co., Ltd; Building 9, 1835 Duhui Road, Minhang Zone, Shanghai 201108, China
| | - Wei Zhang
- School of Pharmacy, Fudan University; 826 Zhangheng Road, Pudong Zone, Shanghai 201203, China
| | - Xinsheng Lei
- School of Pharmacy, Fudan University; 826 Zhangheng Road, Pudong Zone, Shanghai 201203, China
| |
Collapse
|
3
|
Tsalyy A, Kráľ M, Reiberger R, Majer P, Konvalinka J, Kožíšek M, Machara A. Design, synthesis, and activity evaluation of C-8 arylated luteolin derivatives as influenza endonuclease inhibitors. Bioorg Med Chem Lett 2025; 121:130178. [PMID: 40064244 DOI: 10.1016/j.bmcl.2025.130178] [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: 09/17/2024] [Revised: 02/14/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
The polymerase acidic (PA) subunit of the influenza virus, an endonuclease of the RNA-dependent RNA polymerase, represents a viable target for anti-influenza therapies, as evidenced by the efficacy of the FDA-approved drug Xofluza. A characteristic feature of endonuclease inhibitors is their ability to chelate Mg2+ or Mn2+ ions within the enzyme's catalytic site. Previously, our studies identified luteolin and its C-8-glucoside orientin as potent endonuclease inhibitors. This report details our subsequent investigation into the structural modifications of the phenyl moiety attached to the C-8 position of luteolin. The inhibitory potencies (IC50 values) quantified with AlphaScreen technology indicated that substituting the C-8 glucose moiety of orientin resulted in compounds with comparable inhibitory potency. From a series of eighteen compounds, acid 12 with 3-carboxylphenyl moiety at the C-8 position was the most potent inhibitor with nanomolar potency.
Collapse
Affiliation(s)
- Artem Tsalyy
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo n. 2, 166 10 Prague 6, Czech Republic
| | - Michal Kráľ
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo n. 2, 166 10 Prague 6, Czech Republic; First Faculty of Medicine, Charles University, Kateřinská 1660, 121 08 Prague 2, Czech Republic
| | - Róbert Reiberger
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo n. 2, 166 10 Prague 6, Czech Republic; Department of Organic Chemistry, Faculty of Science, Charles University, Hlavova 8, 128 00 Prague 2, Czech Republic
| | - Pavel Majer
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo n. 2, 166 10 Prague 6, Czech Republic
| | - Jan Konvalinka
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo n. 2, 166 10 Prague 6, Czech Republic; Department of Biochemistry, Faculty of Science, Charles University, Hlavova 8, 128 00 Prague 2, Czech Republic
| | - Milan Kožíšek
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo n. 2, 166 10 Prague 6, Czech Republic.
| | - Aleš Machara
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo n. 2, 166 10 Prague 6, Czech Republic.
| |
Collapse
|
4
|
Singh AA, Khan F, Song M. Alleviation of Neurological Disorders by Targeting Neurodegenerative-Associated Enzymes: Natural and Synthetic Molecules. Int J Mol Sci 2025; 26:4707. [PMID: 40429850 PMCID: PMC12112699 DOI: 10.3390/ijms26104707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2025] [Revised: 05/12/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Neurological disorders, encompassing neurodegenerative and neuroinflammatory conditions, present significant public health and clinical challenges. Recent research has elucidated the pivotal role of various enzymes in the onset and progression of these disorders. This review explores the therapeutic potential of targeting these enzymes with natural and synthetic molecules. Key enzymes, including acetylcholinesterase, monoamine oxidase, beta-secretase, tau kinases, caspases, and cyclooxygenase-2, are implicated in diseases such as Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Modulating these enzymes can alleviate symptoms, slow disease progression, or reverse pathological changes. Natural molecules derived from plants, microbes, seaweeds, and animals have long been noted for their therapeutic potential. Their ability to interact with specific enzymes with high specificity and minimal side effects makes them promising candidates for treatment. These natural agents provide a foundation for developing targeted therapies with improved safety profiles. Simultaneously, the development of synthetic chemistry has resulted in molecules designed to inhibit neurodegenerative enzymes with precision. This review examines the progress in creating small molecules, peptides, and enzyme inhibitors through sophisticated drug design techniques. It evaluates the efficacy, safety, and mechanisms of these synthetic agents, highlighting their potential for clinical application. The review offers a comprehensive overview of recent advancements in enzyme-targeted therapies for neurological disorders, covering both natural and synthetic molecules investigated in preclinical and clinical settings. It discusses the mechanisms through which these molecules exert their effects, the challenges faced in their development, and future research directions. By synthesizing current knowledge, this paper aims to illuminate the potential of enzyme-targeted interventions in managing neurological disorders, showcasing both the promise and limitations of these approaches.
Collapse
Affiliation(s)
- Alka Ashok Singh
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Fazlurrahman Khan
- Ocean and Fisheries Development International Cooperation Institute, Pukyong National University, Busan 48513, Republic of Korea
- International Graduate Program of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
5
|
Yan R, Xu YM, Lau ATY. Immobilized metal ion affinity chromatography: waltz of metal ions and biomacromolecules. Expert Rev Proteomics 2025; 22:185-198. [PMID: 40249414 DOI: 10.1080/14789450.2025.2492764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/31/2025] [Accepted: 04/06/2025] [Indexed: 04/19/2025]
Abstract
INTRODUCTION Immobilized metal ion affinity chromatography (IMAC) is an effective method developed in the 1980s for the separation and purification of proteins. The system consists of a solid-phase matrix, a linking ligand, and a metal ion. The method is based on the ability of metal ions to bind specifically to certain specific amino acid residues of proteins, thereby selectively enriching and purifying proteins. AREAS COVERED This review aims to describe current knowledge of fundamental principle of IMAC and summarize the supports, chelating ligands, and metal ions of IMAC. In addition, how IMAC technology is used in proteomics and nucleic acids research are highlighted. EXPERT OPINION Over the past decades, IMAC has been extensively utilized as a predominant technique for protein enrichment in a variety of biological and medical research, such as disease diagnosis, tumor biomarker identification, protein purification, and nucleic acids research. In the future, IMAC should be integrated with other emerging proteomics technologies to promote the applications of metalloproteomes in disease diagnosis, metallodrug development, and clinical translation.
Collapse
Affiliation(s)
- Rui Yan
- The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Yan-Ming Xu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Andy T Y Lau
- The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, People's Republic of China
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| |
Collapse
|
6
|
Faponle AS, Gauld JW, de Visser SP. Insights into Active Site Cysteine Residues in Mycobacterium tuberculosis Enzymes: Potential Targets for Anti-Tuberculosis Intervention. Int J Mol Sci 2025; 26:3845. [PMID: 40332486 PMCID: PMC12028322 DOI: 10.3390/ijms26083845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Cysteine, a semi-essential amino acid, is found in the active site of a number of vital enzymes of the bacterium Mycobacterium tuberculosis (Mtb) and in particular those that relate to its survival, adaptability and pathogenicity. Mtb is the causative agent of tuberculosis, an infectious disease that affects millions of people globally. Common anti-tuberculosis targets are focused on immobilizing a vital cysteine amino acid residue in enzymes that plays critical roles in redox and non-redox catalysis, the modulation of the protein, enzyme activity, protein structure and folding, metal coordination, and posttranslational modifications of newly synthesized proteins. This review examines five Mtb enzymes that contain an active site cysteine residue and are considered as key targets for anti-tuberculosis drugs, namely alkyl hydroperoxide reductase (AhpC), dihydrolipoamide dehydrogenase (Lpd), aldehyde dehydrogenase (ALDH), methionine aminopeptidase (MetAP) and cytochromes P450. AhpC and Lpd protect Mtb against oxidative and nitrosative stress, whereas AhpC neutralizes peroxide/peroxynitrite substrates with two active site cysteine residues. Mtb ALDH detoxifies aldehydes, using a nucleophilic active site cysteine to form an oxyanion thiohemiacetal intermediate, whereas MtMetAP's active site cysteine is essential for substrate recognition. The P450s metabolize various endogenous and exogenous compounds. Targeting these critical active site cysteine residues could disrupt enzyme functions, presenting a promising avenue for developing anti-mycobacterial agents.
Collapse
Affiliation(s)
- Abayomi S. Faponle
- Department of Biochemistry, Faculty of Basic Medical Sciences, Sagamu Campus, Olabisi Onabanjo University, Ago-Iwoye 120107, Nigeria
| | - James W. Gauld
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, ON N9B 3P4, Canada;
- Department of Chemistry, Memorial University of Newfoundland, St. John’s, NL A1C 5S7, Canada
| | - Sam P. de Visser
- Department of Chemical Engineering, Manchester Institute of Biotechnology, The University of Manchester, 131 Princess Street, Manchester M1 7DN, UK
| |
Collapse
|
7
|
Bogacz I, Szilagyi E, Makita H, Simon PS, Zhang M, Doyle MD, Chatterjee K, Kretzschmar M, Chernev P, Croy N, Cheah MH, Dasgupta M, Nangca I, Fransson T, Bhowmick A, Brewster AS, Sauter NK, Owada S, Tono K, Zerdane S, Oggenfuss A, Babich D, Sander M, Mankowsky R, Lemke HT, Gee LB, Sato T, Kroll T, Messinger J, Alonso-Mori R, Bergmann U, Sokaras D, Yachandra VK, Kern J, Yano J. X-ray Absorption Spectroscopy of Dilute Metalloenzymes at X-ray Free-Electron Lasers in a Shot-by-Shot Mode. J Phys Chem Lett 2025; 16:3778-3787. [PMID: 40193717 PMCID: PMC12010424 DOI: 10.1021/acs.jpclett.5c00399] [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: 02/07/2025] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/09/2025]
Abstract
X-ray absorption spectroscopy (XAS) of 3d transition metals provides important electronic structure information for many fields. However, X-ray-induced radiation damage under physiological temperature has prevented using this method to study dilute aqueous systems, such as metalloenzymes, as the catalytic reaction proceeds. Here we present a new approach to enable operando XAS of dilute biological samples and demonstrate its feasibility with K-edge XAS spectra from the Mn cluster in photosystem II and the Fe-S centers in photosystem I. This approach combines highly efficient sample delivery strategies and a robust signal normalization method with high-transmission Bragg diffraction-based spectrometers at X-ray free-electron lasers (XFELs) in a damage-free, shot-by-shot mode. These photon-out spectrometers have been optimized for discriminating the metal Mn/Fe Kα fluorescence signals from the overwhelming scattering background present on currently available detectors for XFELs that lack suitable energy discrimination. We quantify the enhanced performance metrics of the spectrometer and discuss its potential applications for acquiring time-resolved XAS spectra of biological samples during their reactions at XFELs.
Collapse
Affiliation(s)
- Isabel Bogacz
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Erzsi Szilagyi
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
- Department
of Physics, University of Wisconsin−Madison, Madison, Wisconsin 53706, United States
| | - Hiroki Makita
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Philipp S. Simon
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Miao Zhang
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Margaret D. Doyle
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Kuntal Chatterjee
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Moritz Kretzschmar
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
- Department
of Biology, Humboldt-Universität
zu Berlin, D 10099 Berlin, Germany
| | - Petko Chernev
- Molecular
Biomimetics, Department of Chemistry - Ångström, Molecular
Biomimetics, Uppsala University, SE 75120 Uppsala, Sweden
| | - Nicholas Croy
- Molecular
Biomimetics, Department of Chemistry - Ångström, Molecular
Biomimetics, Uppsala University, SE 75120 Uppsala, Sweden
| | - Mun-Hon Cheah
- Molecular
Biomimetics, Department of Chemistry - Ångström, Molecular
Biomimetics, Uppsala University, SE 75120 Uppsala, Sweden
| | - Medhanjali Dasgupta
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Isabela Nangca
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Thomas Fransson
- Department
of Theoretical Chemistry and Biology, KTH
Royal Institute of Technology, 114 28 Stockholm, Sweden
| | - Asmit Bhowmick
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Aaron S. Brewster
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Nicholas K. Sauter
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Shigeki Owada
- Japan Synchrotron
Radiation Research Institute, 1-1-1 Kouto, Sayo-cho, Sayo-gun, Hyogo 679-5198, Japan
- RIKEN
SPring-8 Center, 1-1-1
Kouto, Sayo-cho, Sayo-gun, Hyogo 679-5148, Japan
| | - Kensuke Tono
- Japan Synchrotron
Radiation Research Institute, 1-1-1 Kouto, Sayo-cho, Sayo-gun, Hyogo 679-5198, Japan
- RIKEN
SPring-8 Center, 1-1-1
Kouto, Sayo-cho, Sayo-gun, Hyogo 679-5148, Japan
| | - Serhane Zerdane
- SwissFEL, Paul
Scherrer Institut, 5232 Villigen, Switzerland
| | | | - Danylo Babich
- SwissFEL, Paul
Scherrer Institut, 5232 Villigen, Switzerland
| | - Mathias Sander
- SwissFEL, Paul
Scherrer Institut, 5232 Villigen, Switzerland
| | - Roman Mankowsky
- SwissFEL, Paul
Scherrer Institut, 5232 Villigen, Switzerland
| | - Henrik T. Lemke
- SwissFEL, Paul
Scherrer Institut, 5232 Villigen, Switzerland
| | - Leland B. Gee
- Linac
Coherent Light Source, SLAC National Accelerator
Laboratory, Menlo
Park, California 94025, United States
| | - Takahiro Sato
- Linac
Coherent Light Source, SLAC National Accelerator
Laboratory, Menlo
Park, California 94025, United States
| | - Thomas Kroll
- SSRL, SLAC National Accelerator Laboratory, Menlo Park, California 94025, United States
| | - Johannes Messinger
- Molecular
Biomimetics, Department of Chemistry - Ångström, Molecular
Biomimetics, Uppsala University, SE 75120 Uppsala, Sweden
- Department
of Plant Physiology, Umeå Plant Science Centre, Umeå University, 90187 Umeå, Sweden
| | - Roberto Alonso-Mori
- Linac
Coherent Light Source, SLAC National Accelerator
Laboratory, Menlo
Park, California 94025, United States
| | - Uwe Bergmann
- Department
of Physics, University of Wisconsin−Madison, Madison, Wisconsin 53706, United States
| | - Dimosthenis Sokaras
- SSRL, SLAC National Accelerator Laboratory, Menlo Park, California 94025, United States
| | - Vittal K. Yachandra
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Jan Kern
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Junko Yano
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| |
Collapse
|
8
|
Zhang J, Zhang Q, Ge R, Liu A, Chen B, Zhang Z, Zhao B, Yu J, Zhao Y, Yu L, Cao M, Wang B, Huang X. Enantiodivergent Radical Alkylation by Synergistic Lewis-Acid-Enzyme and Photoredox Catalysis. Angew Chem Int Ed Engl 2025:e202500338. [PMID: 40208083 DOI: 10.1002/anie.202500338] [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: 01/06/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/11/2025]
Abstract
Artificial metalloenzymes (ArMs) and photoenzymatic catalysis represent two cutting-edge approaches to creating new enzyme reactivity. However, the potential of merging these two strategies remains underdeveloped for enantiocontrolled biotransformations. Herein, we develop a synergistic metalloenzymatic and photoredox catalysis platform to enable enantiodivergent radical alkylation of 2-acyl imidazoles. Specifically, cupin proteins are redesigned to function as copper(II)-based Lewis-acid-enzymes (LAses), which, in synergy with tripyridinyl-ruthenium-based photoredox catalysis, precisely control the generation, reactivity, and selectivity of abiological radicals, thereby unlocking non-natural enzyme reactivity. Powered by protein engineering, repurposed photo-LAses facilitate the green and efficient synthesis of diverse enantioenriched α-chiral ketones in high enantioselectivity (both enantiomers accessible, up to 97% yield and 98.5:1.5 enantiomeric ratio [er]). Detailed mechanistic studies suggest a radical addition to the metalloenzymatic enolate pathway and explain the switched selectivity from dark to photoconditions.
Collapse
Affiliation(s)
- Jiawei Zhang
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), Frontier Interdisciplinary Science Research Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P.R. China
| | - Qiaoyu Zhang
- State Key Laboratory of Physical Chemistry of Solid Surfaces and Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, P.R. China
| | - Ran Ge
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Key Laboratory for Synthetic Biotechnology of Xiamen City, Xiamen University, Xiamen, 361005, P.R. China
| | - Aokun Liu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, P.R. China
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, P.R. China
| | - Bin Chen
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), Frontier Interdisciplinary Science Research Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P.R. China
| | - Zihan Zhang
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), Frontier Interdisciplinary Science Research Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P.R. China
| | - Beibei Zhao
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), Frontier Interdisciplinary Science Research Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P.R. China
| | - Jinhai Yu
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), Frontier Interdisciplinary Science Research Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P.R. China
| | - Yue Zhao
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), Frontier Interdisciplinary Science Research Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P.R. China
| | - Lu Yu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, P.R. China
| | - Mingfeng Cao
- Department of Chemical and Biochemical Engineering, College of Chemistry and Chemical Engineering, Key Laboratory for Synthetic Biotechnology of Xiamen City, Xiamen University, Xiamen, 361005, P.R. China
| | - Binju Wang
- State Key Laboratory of Physical Chemistry of Solid Surfaces and Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, P.R. China
| | - Xiaoqiang Huang
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), Frontier Interdisciplinary Science Research Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P.R. China
| |
Collapse
|
9
|
Nikopaschou M, Samiotaki M, Stylianaki EA, Król K, Gragera P, Raja A, Aidinis V, Chroni A, Fruci D, Panayotou G, Stratikos E. ERAP1 Activity Modulates the Immunopeptidome but Also Affects the Proteome, Metabolism, and Stress Responses in Cancer Cells. Mol Cell Proteomics 2025; 24:100964. [PMID: 40189142 DOI: 10.1016/j.mcpro.2025.100964] [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: 09/22/2024] [Revised: 03/23/2025] [Accepted: 04/02/2025] [Indexed: 05/11/2025] Open
Abstract
Endoplasmic reticulum (ER) aminopeptidase 1 (ERAP1) metabolizes peptides inside the ER and shapes the peptide repertoire available for binding to major histocompatibility complex class I molecules (MHC-I). However, it may have additional effects on cellular homeostasis, which have not been explored. To address these questions, we used both genetic silencing of ERAP1 expression as well as treatment with a selective allosteric ERAP1 inhibitor to probe changes in the immunopeptidome and proteome of the A375 melanoma cancer cell line. We observed significant immunopeptidome shifts with both methods of functional ERAP1 disruption, which were distinct for each method. Both methods of inhibition led to an enhancement, albeit slight, in tumor cell killing by stimulated human peripheral blood mononuclear cells and in significant proteomic alterations in pathways related to metabolism and cellular stress. Similar proteomic changes were also observed in the leukemia cell line THP-1. Biochemical analyses suggested that ERAP1 inhibition affected sensitivity to ER stress, reactive oxygen species production, and mitochondrial metabolism. Although the proteomics shifts were significant, their potential in shaping immunopeptidome shifts was limited since only 9.6% of differentially presented peptides belonged to proteins with altered expression and only 4.0% of proteins with altered expression were represented in the immunopeptidome shifts. Taken together, our findings suggest that modulation of ERAP1 activity can generate unique immunopeptidomes, mainly due to altered peptide processing in the ER, but also induce changes in the cellular proteome and metabolic state which may have further effects on tumor cells.
Collapse
Affiliation(s)
- Martha Nikopaschou
- National Centre for Scientific Research Demokritos, Agia Paraskevi, Greece; Department of Chemistry, National and Kapodistrian University of Athens, Zografou, Greece
| | - Martina Samiotaki
- Biomedical Sciences Research Center "Alexander Fleming", Institute for Bioinnovation, Vari, Greece
| | - Elli-Anna Stylianaki
- Biomedical Sciences Research Center "Alexander Fleming", Institute for Fundamental Biomedical Research, Vari, Greece
| | - Kamila Król
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Paula Gragera
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Aroosha Raja
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Vassilis Aidinis
- Biomedical Sciences Research Center "Alexander Fleming", Institute for Fundamental Biomedical Research, Vari, Greece
| | - Angeliki Chroni
- National Centre for Scientific Research Demokritos, Agia Paraskevi, Greece
| | - Doriana Fruci
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - George Panayotou
- Biomedical Sciences Research Center "Alexander Fleming", Institute for Bioinnovation, Vari, Greece
| | - Efstratios Stratikos
- National Centre for Scientific Research Demokritos, Agia Paraskevi, Greece; Department of Chemistry, National and Kapodistrian University of Athens, Zografou, Greece.
| |
Collapse
|
10
|
Wu Y, Li J, Jiang W, Xu W, Zheng L, Wang C, Gu W, Zhu C. Second coordination sphere regulates nanozyme inhibition to assist early drug discovery. Nat Commun 2025; 16:3123. [PMID: 40169567 PMCID: PMC11961690 DOI: 10.1038/s41467-025-58291-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 03/17/2025] [Indexed: 04/03/2025] Open
Abstract
Early drug discovery is a time- and cost-consuming task requiring enzymes. Although nanozymes with metal sites akin to metallocofactors display similar activities, the lack of proximal amino acids hinders them from more adequately mimicking enzymes for drug discovery purposes. Hence, the rational design of the nanozyme second coordination sphere is desirable yet remains challenging. Herein, we report a nanozyme featuring atomically dispersed Cu-N4 sites with proximal hydroxyl groups (CuNC-OH). Experimental and theoretical results reveal that Cu-N4 site and hydroxyl respectively behave as cofactor and amino acid of the enzymatic pocket to interact with adsorbates, regulating nanozyme activity and inhibition. This mechanism involving dual sites is similar to that of thyroid peroxidases, which enables specific inhibition of CuNC-OH by antithyroid drugs. Based on these findings, a nanozyme-assisted drug discovery kit is established to analyze inhibition features of thyroid peroxidase inhibitors and screen out promising antithyroid drugs with a significant cost reduction compared with traditional enzyme kits.
Collapse
Affiliation(s)
- Yu Wu
- State Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, PR China
| | - Jian Li
- State Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, PR China
| | - Wenxuan Jiang
- State Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, PR China
| | - Weiqing Xu
- State Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, PR China
| | - Lirong Zheng
- Beijing Synchrotron Radiation Facility, Chinese Academy of Science, Beijing, PR China
| | - Canglong Wang
- Institute of Modern Physics, Chinese Academy of Science, Lanzhou, PR China
| | - Wenling Gu
- State Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, PR China
| | - Chengzhou Zhu
- State Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan, PR China.
| |
Collapse
|
11
|
Shirazi J, Jafari S, Ryde U, Irani M. Metal preference of glyoxalase II, a quantum mechanics/molecular mechanics study. Dalton Trans 2025; 54:5779-5795. [PMID: 40066953 DOI: 10.1039/d4dt03519d] [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: 04/02/2025]
Abstract
The catalytic activity of the binuclear glyoxalase II (GlxII) enzyme is closely linked to the type and charge of metal ions in its active site. Using hybrid quantum mechanics/molecular mechanics (QM/MM) calculations, we investigated the reaction mechanism of human GlxII, which features two Zn(II) ions in its active site. By systematically replacing these Zn(II) ions with Fe(II), Fe(III), or Co(II), we evaluated the impact of metal substitutions on reaction energetics and active-site geometry. Our results reveal that the type and position of the metal ions are critical to the catalytic activity of GlxII. Substitution of the Zn(II) ion in the three-histidine site with Fe(II), Fe(III), or Co(II) significantly increased the activation barrier, indicating that these configurations are less favorable. In contrast, substituting Zn(II) in the two-histidine site with either Fe(II) or Co(II) resulted in a reduced activation barrier and produced geometries closely resembling those observed when both metal sites are occupied by Zn(II). Additionally, moving the metal ions from the QM to the MM region inhibited the reaction, highlighting their direct chemical involvement in catalysis beyond electrostatic stabilization. These results underscore that the metal ions chemically participate in the catalytic process beyond their electrostatic contributions. Collectively, our results provide insights into the structural and electronic factors governing GlxII catalysis, offering a theoretical framework to complement and refine experimental studies.
Collapse
Affiliation(s)
- Javad Shirazi
- Department of Chemistry, University of Kurdistan, Sanandaj 66177-15175, Iran.
| | - Sonia Jafari
- Department of Chemistry, University of Kurdistan, Sanandaj 66177-15175, Iran.
| | - Ulf Ryde
- Department of Theoretical Chemistry, Lund University, P.O. Box 124, SE-221 00 Lund, Sweden
| | - Mehdi Irani
- Department of Chemistry, University of Kurdistan, Sanandaj 66177-15175, Iran.
| |
Collapse
|
12
|
Zhou Y, Wang J, Sun Y, Cheng Y, Wu W. Non-Hydroxamate Inhibitors of IspC Enzyme in the MEP Pathway: Structural Insights and Drug Development Potential. Chem Biol Drug Des 2025; 105:e70086. [PMID: 40099748 DOI: 10.1111/cbdd.70086] [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: 11/13/2024] [Revised: 02/23/2025] [Accepted: 02/27/2025] [Indexed: 03/20/2025]
Abstract
1-Deoxy-D-xylulose-5-phosphate reductoisomerase (IspC) is a key enzyme in the MEP pathway, essential for many bacteria, human pathogens, and plants, thus being an attractive drug target. Fosmidomycin, a potent IspC inhibitor with hydroxamate metal-binding pharmacophores (MBPs), has entered clinical trials for malaria but is hampered by pharmacokinetic and toxicity issues of the hydroxamate fragment. This has led to increased interest in non-hydroxamate inhibitors. This review focuses on the crystal structure and active-site binding mode of IspC, and the structural types, inhibitory activities, and structure-activity relationships of non-hydroxamate IspC inhibitors. Early attempts to design such inhibitors involved direct removal or replacement of the hydroxamate MBPs, with varying results. Lipophilic inhibitors, bisubstrate inhibitors, and those developed for herbicidal applications have shown promise. However, challenges remain due to the sensitivity of the enzyme active site to ligand interactions. Future research could draw from other metalloenzyme studies to develop novel and efficient non-hydroxamate IspC inhibitors.
Collapse
Affiliation(s)
- Yaqing Zhou
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| | - Jili Wang
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| | - Yong Sun
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| | - Yarui Cheng
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| | - Wenhai Wu
- Shiyan Key Laboratory of Biological Resources and eco-Environmental Protection, College of Chemical and Environmental Engineering, Hanjiang Normal University, Shiyan, China
| |
Collapse
|
13
|
Meegan MJ. Feature Reviews in Medicinal Chemistry. Pharmaceuticals (Basel) 2025; 18:260. [PMID: 40006073 PMCID: PMC11859477 DOI: 10.3390/ph18020260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
This Special Issue of Pharmaceuticals ("Feature reviews in Medicinal Chemistry") contains a series of reviews covering a broad selection of topics in Medicinal Chemistry, which are chosen to illustrate important recent results in this fast-evolving interdisciplinary research area that aims to improve human health by developing drugs to combat diseases [...].
Collapse
Affiliation(s)
- Mary J Meegan
- School of Pharmacy and Pharmaceutical Sciences, Panoz Institute, Trinity College Dublin, D02 PN40 Dublin, Ireland
| |
Collapse
|
14
|
Kráľ M, Kotačka T, Reiberger R, Panýrková G, Radilová K, Osifová Z, Flieger M, Konvalinka J, Majer P, Kožíšek M, Machara A. 3'-Dehydroxypurpurogallin-4-Carboxamides as Influenza A Endonuclease Inhibitors: Synthesis, Structure-Activity Relationship Analysis, and Structural Characterization of Protein Complex. ChemMedChem 2025; 20:e202400577. [PMID: 39400442 DOI: 10.1002/cmdc.202400577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/07/2024] [Accepted: 10/07/2024] [Indexed: 10/15/2024]
Abstract
The influenza RNA-dependent RNA polymerase harbours an endonuclease subunit characterized by a catalytic site housing two divalent metal ions. By effectively chelating both Mg2+ and Mn2+ ions, a small-molecule inhibitor with a metal-binding pharmacophore can halt endonuclease activity. Herein, two 3'-dehydroxypurpurogallin-4-carboxamide series, namely twelve C-4' unsubstituted and twelve C-4' phenyl substituted congeners were designed and prepared to be tested as inhibitors of the metal-dependent viral enzyme. These inhibitors were accessed through the chemoenzymatic reaction of gallic acid with either pyrocatechol or phenylpyrocatechol moderated by laccase, followed by amidation. Experimental IC50 values were determined using AlphaScreen technology, with the most potent inhibitors exhibiting IC50 values around 0.35 μM. Using X-ray crystallography, we analyzed structure of the endonuclease in complex with one potent 3'-dehydroxypurpurogallin-carboxamide at 2.0 Å resolution, revealing the coordination of the compound's triad of oxygen atoms with the two metal ions in the influenza A endonuclease active site.
Collapse
Affiliation(s)
- Michal Kráľ
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 542/2, Prague 6, 160 00, Czech Republic
- First Faculty of Medicine, Charles University in Prague, Kateřinská 1660/32, Prague 2, 121 08, Czech Republic
| | - Tomáš Kotačka
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 542/2, Prague 6, 160 00, Czech Republic
- First Faculty of Medicine, Charles University in Prague, Kateřinská 1660/32, Prague 2, 121 08, Czech Republic
| | - Róbert Reiberger
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 542/2, Prague 6, 160 00, Czech Republic
- Department of Organic Chemistry, Faculty of Science, Charles University in Prague, Hlavova 2030/8, Prague 2, 128 43, Czech Republic
| | - Gabriela Panýrková
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 542/2, Prague 6, 160 00, Czech Republic
| | - Kateřina Radilová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 542/2, Prague 6, 160 00, Czech Republic
| | - Zuzana Osifová
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 542/2, Prague 6, 160 00, Czech Republic
- Department of Organic Chemistry, Faculty of Science, Charles University in Prague, Hlavova 2030/8, Prague 2, 128 43, Czech Republic
| | - Miroslav Flieger
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 4, 140 00, Czech Republic
| | - Jan Konvalinka
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 542/2, Prague 6, 160 00, Czech Republic
| | - Pavel Majer
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 542/2, Prague 6, 160 00, Czech Republic
| | - Milan Kožíšek
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 542/2, Prague 6, 160 00, Czech Republic
| | - Aleš Machara
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo náměstí 542/2, Prague 6, 160 00, Czech Republic
| |
Collapse
|
15
|
Xiong G, Xiao Z. Computational approaches for the identification of novel metal-binding pharmacophores: advances and challenges. Drug Discov Today 2025; 30:104293. [PMID: 39805538 DOI: 10.1016/j.drudis.2025.104293] [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: 11/07/2024] [Revised: 01/02/2025] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Metalloenzymes are important therapeutic targets for a variety of human diseases. Computational approaches have recently emerged as effective tools to understand metal-ligand interactions and expand the structural diversity of both metalloenzyme inhibitors (MIs) and metal-binding pharmacophores (MBPs). In this review, we highlight key advances in currently available fine-tuning modeling methods and data-driven cheminformatic approaches. We also discuss major challenges to the recognition of novel MBPs and MIs. The evidence provided herein could expedite future computational efforts to guide metalloenzyme-based drug discovery.
Collapse
Affiliation(s)
- Guoli Xiong
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhiyan Xiao
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
16
|
Jin C, Xiu WL, Liu Y, Hao XW, Dong Q. Clinical features and outcomes of pseudolithiasis induced by ceftriaxone in Chinese children: a single-center observational study. Front Pediatr 2025; 13:1527014. [PMID: 39896723 PMCID: PMC11782244 DOI: 10.3389/fped.2025.1527014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/02/2025] [Indexed: 02/04/2025] Open
Abstract
INTRODUCTION Ceftriaxone (CTX) is widely used in pediatric infectious disease treatment, although the diagnostic and therapeutic management of CTX-induced gallbladder pseudolithiasis (PL) remains challenging. In this study, we investigated the occurrence, clinical features, and management of CTX-induced PL in children. METHODS A retrospective case-control study was conducted on 185 pediatric patients receiving CTX at a single center. Data on treatment regimens, gallbladder imaging findings, and serum biochemical parameters post-CTX therapy were analyzed. Patients were classified into PL (n = 34) and non-PL (n = 151) groups based on imaging findings. RESULTS PL was diagnosed in 18.4% of patients treated with CTX, primarily through ultrasound, which revealed hyperechoic material within the gallbladder. Compared with the non-PL group, patients with PL were older and taller, with no significant differences in CTX dosage (p = 0.915). Patients with PL also had higher rates of digestive and neurological infections (both p < 0.001). Serum analysis revealed distinct liver and kidney function markers in the PL group, including lower levels of total bile acids, adenosine deaminase, and lactate dehydrogenase, and higher creatinine levels (all p < 0.05). Discontinuation of CTX led to symptom resolution in most cases, and all cases of PL resolved within three months. CONCLUSIONS The occurrence of PL is not significantly related to CTX dosage. Furthermore, the rate of CTX metabolism and excretion may play a key role in PL development. Overall, the findings demonstrate that ultrasound is an effective tool for monitoring the development of PL in children receiving CTX and that discontinuation of CTX could be an effective treatment for PL.
Collapse
Affiliation(s)
- Chen Jin
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Shandong Provincial Key Laboratory of Digital Medicine and Computer-Assisted Surgery, Qingdao University, Qingdao, Shandong, China
| | - Wen-Li Xiu
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Shandong Provincial Key Laboratory of Digital Medicine and Computer-Assisted Surgery, Qingdao University, Qingdao, Shandong, China
| | - Yao Liu
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Shandong Provincial Key Laboratory of Digital Medicine and Computer-Assisted Surgery, Qingdao University, Qingdao, Shandong, China
| | - Xi-Wei Hao
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Qian Dong
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
- Shandong Provincial Key Laboratory of Digital Medicine and Computer-Assisted Surgery, Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
17
|
Lee H, Diao J, Tian Y, Guleria R, Lee E, Smith A, Savage M, Yeh D, Roberson L, Blenner M, Tang YJ, Moon TS. Developing an alternative medium for in-space biomanufacturing. Nat Commun 2025; 16:728. [PMID: 39819985 PMCID: PMC11739595 DOI: 10.1038/s41467-025-56088-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 01/08/2025] [Indexed: 01/19/2025] Open
Abstract
In-space biomanufacturing provides a sustainable solution to facilitate long-term, self-sufficient human habitation in extraterrestrial environments. However, its dependence on Earth-supplied feedstocks renders in-space biomanufacturing economically nonviable. Here, we develop a process termed alternative feedstock-driven in-situ biomanufacturing (AF-ISM) to alleviate dependence on Earth-based resupply of feedstocks. Specifically, we investigate three alternative feedstocks (AF)-Martian and Lunar regolith, post-consumer polyethylene terephthalate, and fecal waste-to develop an alternative medium for lycopene production using Rhodococcus jostii PET strain S6 (RPET S6). Our results show that RPET S6 could directly utilize regolith simulant particles as mineral replacements, while the addition of anaerobically pretreated fecal waste synergistically supported its cell growth. Additionally, lycopene production using AF under microgravity conditions achieved levels comparable to those on Earth. Furthermore, an economic analysis shows significant lycopene production cost reductions using AF-ISM versus conventional methods. Overall, this work highlights the viability of AF-ISM for in-space biomanufacturing.
Collapse
Affiliation(s)
- Hakyung Lee
- Washington University in St. Louis, Saint Louis, MO, USA
| | - Jinjin Diao
- Washington University in St. Louis, Saint Louis, MO, USA.
| | - Yuxin Tian
- Washington University in St. Louis, Saint Louis, MO, USA
- Synthetic Biology Group, J. Craig Venter Institute, La Jolla, CA, USA
| | | | - Eunseo Lee
- Washington University in St. Louis, Saint Louis, MO, USA
| | | | - Millie Savage
- Lincoln University of Missouri, Jefferson City, MO, USA
| | - Daniel Yeh
- University of South Florida, Tampa, FL, USA
| | - Luke Roberson
- National Aeronautics and Space Administration, John F. Kennedy Space Center, Merritt Island, FL, USA
| | | | - Yinjie J Tang
- Washington University in St. Louis, Saint Louis, MO, USA.
| | - Tae Seok Moon
- Washington University in St. Louis, Saint Louis, MO, USA.
- Synthetic Biology Group, J. Craig Venter Institute, La Jolla, CA, USA.
| |
Collapse
|
18
|
Viktorova VV, Obydennov DL, Kovaleva KS, Yarovaya OI, Khasanov SA, Bormotov NI, Esaulkova IL, Serova OA, Zarubaev VV, Shishkina LN, Salakhutdinov NF, Sosnovskikh VY. The Reaction of Fenchone and Camphor Hydrazones with 5-Acyl-4-Pyrones as a Method for the Synthesis of New Polycarbonyl Conjugates: Tautomeric Equilibrium and Antiviral Activity. Chem Biodivers 2025; 22:e202401461. [PMID: 39233581 DOI: 10.1002/cbdv.202401461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/06/2024]
Abstract
Selective synthesis of polycarbonyl conjugates of (+)-fenchone and (+)-camphor was carried out (44-91 % yields) via the ring-opening transformation of 5-acyl-4-pyrones with hydrazones of the corresponding monoterpenoids. A strong influence of the hydrazone fragment on the observed tautomeric equilibrium of the tricarbonyl system was shown. Although the major tautomer of the conjugates is the acyclic polycarbonyl form, the camphor-based conjugates undergo new type of ring-chain tautomerism, diketoenaminone-dihydropyridone equilibrium, and predominantly exist in the cyclic dihydropyridone form in DMSO-d6. The polyketones can undergo intramolecular cyclization to form N-amino-4-pyridones in high selectivity. In vitro screening for activity against the influenza virus H1 N1 and vaccinia virus was estimated for the obtained conjugates. The (+)-fenchone derivatives demonstrated the higher activity against vaccinia virus than camphor derivatives. The conjugate, which was prepared from diethyl isochelidonate and hydrazone (+)-fenchone, showed the highest activity against vaccinia virus (SI=17).
Collapse
Affiliation(s)
- Viktoria V Viktorova
- Institute of Natural Sciences and Mathematics, Ural Federal University, 620000, Ekaterinburg, Russian Federation
| | - Dmitrii L Obydennov
- Institute of Natural Sciences and Mathematics, Ural Federal University, 620000, Ekaterinburg, Russian Federation
| | - Kseniya S Kovaleva
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, 630090, Novosibirsk, Russian Federation
| | - Olga I Yarovaya
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, 630090, Novosibirsk, Russian Federation
| | | | - Nikolay I Bormotov
- State Research Center of Virology and Biotechnology "VECTOR", Novosibirsk Region, 6300559, Koltsovo, Russian Federation
| | - Iana L Esaulkova
- St. Petersburg Pasteur Institute, 197001, St. Petersburg, Russian Federation
| | - Olga A Serova
- State Research Center of Virology and Biotechnology "VECTOR", Novosibirsk Region, 6300559, Koltsovo, Russian Federation
| | - Vladimir V Zarubaev
- St. Petersburg Pasteur Institute, 197001, St. Petersburg, Russian Federation
| | - Larisa N Shishkina
- State Research Center of Virology and Biotechnology "VECTOR", Novosibirsk Region, 6300559, Koltsovo, Russian Federation
| | - Nariman F Salakhutdinov
- N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, 630090, Novosibirsk, Russian Federation
| | - Vyacheslav Y Sosnovskikh
- Institute of Natural Sciences and Mathematics, Ural Federal University, 620000, Ekaterinburg, Russian Federation
| |
Collapse
|
19
|
Gao S, Hou Y, Xu Y, Li J, Zhang C, Jiang S, Yu S, Liu L, Tu W, Yu B, Zhang Y, Li L. Discovery of orally bioavailable phosphonate prodrugs of potent ENPP1 inhibitors for cancer treatment. Eur J Med Chem 2024; 279:116853. [PMID: 39270452 DOI: 10.1016/j.ejmech.2024.116853] [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: 05/23/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024]
Abstract
Ectonucleotide pyrophosphatase phosphodiesterase 1 (ENPP1) is the dominant hydrolase of 2',3'-cyclic GMP-AMP (cGAMP). Inhibition of ENPP1 contributes to increased cGAMP concentration and stimulator of interferon gene (STING) activation, with the potential to boost immune response against cancer. ENPP1 is a promising therapeutic target in tumor immunotherapy. To date, orally bioavailable ENPP1 inhibitors with highly potent activity under physiological conditions have been rarely reported. Herein, we report our effort in the design and synthesis of two different series of ENPP1 inhibitors, and in the identification of a highly potent ENPP1 inhibitor 27 (IC50 = 1.2 nM at pH 7.5), which significantly enhanced the cGAMP-mediated STING activity in THP-1 cells. Phosphonate compound 27 has good preclinical pharmacokinetic profiles with low plasma clearance rate in mouse, rat, and dog. It has been developed as bis-POM prodrug 36 which successfully improves the oral bioavailability of 27. In the Pan02 syngeneic mouse model of pancreatic cancer, orally administered 36 showed synergistic effect in combination with radiotherapy.
Collapse
Affiliation(s)
- Shanyun Gao
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865, Zuchongzhi Road, Zhangjiang Science City, Shanghai, 201203, China
| | - Yingjie Hou
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865, Zuchongzhi Road, Zhangjiang Science City, Shanghai, 201203, China
| | - Yanxiao Xu
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865, Zuchongzhi Road, Zhangjiang Science City, Shanghai, 201203, China
| | - Jingjing Li
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865, Zuchongzhi Road, Zhangjiang Science City, Shanghai, 201203, China
| | - Chaobo Zhang
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865, Zuchongzhi Road, Zhangjiang Science City, Shanghai, 201203, China
| | - Shujuan Jiang
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865, Zuchongzhi Road, Zhangjiang Science City, Shanghai, 201203, China
| | - Songda Yu
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865, Zuchongzhi Road, Zhangjiang Science City, Shanghai, 201203, China
| | - Lei Liu
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865, Zuchongzhi Road, Zhangjiang Science City, Shanghai, 201203, China
| | - Wangyang Tu
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865, Zuchongzhi Road, Zhangjiang Science City, Shanghai, 201203, China.
| | - Bing Yu
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865, Zuchongzhi Road, Zhangjiang Science City, Shanghai, 201203, China.
| | - Yixiang Zhang
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865, Zuchongzhi Road, Zhangjiang Science City, Shanghai, 201203, China.
| | - Leping Li
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865, Zuchongzhi Road, Zhangjiang Science City, Shanghai, 201203, China.
| |
Collapse
|
20
|
Huo X, Zhao F, Xu Y, Liu Q, Wang W, Yang C, Su J. Fabulous combination therapy: Synergistic antibiotic inhibition of aquatic antibiotic-resistant bacteria via membrane damage and DNA binding by novel nano antimicrobial peptide C-I20. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136225. [PMID: 39442310 DOI: 10.1016/j.jhazmat.2024.136225] [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: 07/20/2024] [Revised: 10/01/2024] [Accepted: 10/19/2024] [Indexed: 10/25/2024]
Abstract
Aquatic microbiota' antibiotic resistance undermines traditional treatment efficacy, posing a severe threat to sustainable water environment. Our study addresses this challenge through a fantastic approach involving novel nano antimicrobial peptide C-I20 and antibiotics. Antibacterial tests demonstrated that C-I20 effectively combated both standard and aquatic pathogenic resistant strains. C-I20 killed drug-resistant bacteria by disrupting membrane structure and binding to DNA. C-I20 bound to DNA, forming precipitates susceptible to rapid degradation by trypsin and DNase I. When combined with chloramphenicol, florfenicol, ampicillin, or enrofloxacin, C-I20 exhibited remarkably higher inhibitory rates against bacteria compared to individual use of C-I20 or antibiotics alone. Continuous passage analysis revealed that co-administration of C-I20 with chloramphenicol, florfenicol, ampicillin, and enrofloxacin delays the emergence and progression of antibiotic resistance. This combination therapy was proved to be highly effective, notably reducing tissue bacterial loads and pathological changes. Evaluation in an Aeromonas hydrophila infection model showed the lowest morbidity rate and bacterial loading in the C-I20 combined with ampicillin group. Antimicrobial susceptibility analysis confirmed that C-I20 supplementation markedly suppresses ampicillin-induced intestinal resistant bacteria. In conclusion, C-I20 in conjunction with antibiotic therapy effectively inhibits infection and drug-resistant bacterial development, offering a promising strategy for managing drug-resistant bacteria in aquatic animals.
Collapse
Affiliation(s)
- Xingchen Huo
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Fengxia Zhao
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Yuezong Xu
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Qian Liu
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Weicheng Wang
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Chunrong Yang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Jianguo Su
- Hubei Hongshan Laboratory, College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, China.
| |
Collapse
|
21
|
Baumann A, Papenkordt N, Robaa D, Szigetvari PD, Vogelmann A, Bracher F, Sippl W, Jung M, Haavik J. Aromatic Amino Acid Hydroxylases as Off-Targets of Histone Deacetylase Inhibitors. ACS Chem Neurosci 2024; 15:4143-4155. [PMID: 39523540 PMCID: PMC11587510 DOI: 10.1021/acschemneuro.4c00346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 10/26/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
The aromatic amino acid hydroxylases (AAAHs) phenylalanine hydroxylase, tyrosine hydroxylase, and tryptophan hydroxylases 1 and 2 are structurally related enzymes that contain an active site iron atom and depend on tetrahydrobiopterin (BH4) as cosubstrate. Due to their important roles in synthesis of serotonin, dopamine, noradrenaline, and adrenaline and their involvement in cardiovascular, neurological, and endocrine disorders, AAAHs have been targeted by substrate analogs, iron chelators, and allosteric ligands. Phenylalanine hydroxylase is also off-target of the histone deacetylase (HDAC) inhibitor panobinostat. To systematically explore the binding of HDAC inhibitors to AAAHs, we screened a library of 307 HDAC inhibitors and structural analogs against tryptophan hydroxylase 1 using a fluorescence-based thermal stability assay, followed by activity assays. Selected hits were enzymatically tested against all four purified human AAAHs. Cellular thermal shift assay was performed for phenylalanine hydroxylase. We show that panobinostat and structurally related compounds such as TB57, which similarly to panobinostat also contains a cinnamoyl hydroxamate, bind to human AAAHs and inhibit these enzymes with high selectivity within the class (panobinostat inhibition (IC50): phenylalanine hydroxylase (18 nM) > tyrosine hydroxylase (450 nM) > tryptophan hydroxylase 1 (1960 nM). This study shows that panobinostat and related hydroxamic acid type HDAC inhibitors inhibit all AAAHs at therapeutically relevant concentrations. Our results warrant further investigations of the off-target relevance of HDAC inhibitors intended for clinical use and provide directions for new dual HDAC/AAAH and selective AAAH inhibitors. These findings may also provide a new mechanistic link between regulation of histone modification, AAAH function, and monoaminergic neurotransmission.
Collapse
Affiliation(s)
- Anne Baumann
- Department
of Biomedicine, University of Bergen, 5007 Bergen, Norway
| | - Niklas Papenkordt
- Institute
of Pharmaceutical Sciences, University of
Freiburg, 79104 Freiburg, Germany
| | - Dina Robaa
- Institute
of Pharmacy, Martin-Luther University of
Halle – Wittenberg, 06120 Halle/Saale, Germany
| | - Peter D. Szigetvari
- Department
of Biomedicine, University of Bergen, 5007 Bergen, Norway
- Division
of Psychiatry, Haukeland University Hospital, 5009 Bergen, Norway
| | - Anja Vogelmann
- Institute
of Pharmaceutical Sciences, University of
Freiburg, 79104 Freiburg, Germany
| | - Franz Bracher
- Department
of Pharmacy – Center for Drug Research, Ludwig-Maximilians University Munich, 81377 Munich, Germany
| | - Wolfgang Sippl
- Institute
of Pharmacy, Martin-Luther University of
Halle – Wittenberg, 06120 Halle/Saale, Germany
| | - Manfred Jung
- Institute
of Pharmaceutical Sciences, University of
Freiburg, 79104 Freiburg, Germany
| | - Jan Haavik
- Department
of Biomedicine, University of Bergen, 5007 Bergen, Norway
- Bergen Center
for Brain Plasticity, Division of Psychiatry, Haukeland University Hospital, 5009 Bergen, Norway
| |
Collapse
|
22
|
Zhang L, Ke D, Li Y, Zhang H, Zhang X, Wang S, Ni S, Peng B, Zeng H, Hou T, Du Y, Pan P, Yu Y, Chen W. Design and synthesis of 7-membered lactam fused hydroxypyridinones as potent metal binding pharmacophores (MBPs) for inhibiting influenza virus PA N endonuclease. Eur J Med Chem 2024; 276:116639. [PMID: 38964259 DOI: 10.1016/j.ejmech.2024.116639] [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: 04/28/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/06/2024]
Abstract
Since influenza virus RNA polymerase subunit PAN is a dinuclear Mn2+ dependent endonuclease, metal-binding pharmacophores (MBPs) with Mn2+ coordination has been elucidated as a promising strategy to develop PAN inhibitors for influenza treatment. However, few attentions have been paid to the relationship between the optimal arrangement of the donor atoms in MBPs and anti-influenza A virus (IAV) efficacy. Given that, the privileged hydroxypyridinones fusing a seven-membered lactam ring with diverse side chains, chiral centers or cyclic systems were designed and synthesized. A structure-activity relationship study resulted in a hit compound 16l (IC50 = 2.868 ± 0.063 μM against IAV polymerase), the seven-membered lactam ring of which was fused a pyrrolidine ring. Further optimization of the hydrophobic binding groups on 16l afforded a lead compound (R, S)-16s, which exhibited a 64-fold more potent inhibitory activity (IC50 = 0.045 ± 0.002 μM) toward IAV polymerase. Moreover, (R, S)-16s demonstrated a potent anti-IAV efficacy (EC50 = 0.134 ± 0.093 μM) and weak cytotoxicity (CC50 = 15.35 μM), indicating the high selectivity of (R, S)-16s. Although the lead compound (R, S)-16s exhibited a little weaker activity than baloxavir, these findings illustrated the utility of a metal coordination-based strategy in generating novel MBPs with potent anti-influenza activity.
Collapse
Affiliation(s)
- Lei Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Di Ke
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601, China
| | - Yuting Li
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Hui Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Xi Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Jinhua Institute of Zhejiang University, Jinhua, Zhejiang, 321299, China
| | - Sihan Wang
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Shaokai Ni
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Bo Peng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Huixuan Zeng
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Tingjun Hou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yushen Du
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Peichen Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yongping Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Jinhua Institute of Zhejiang University, Jinhua, Zhejiang, 321299, China; School of Pharmacy, Xinjiang Medical University, Urumqi, 830054, China
| | - Wenteng Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Jinhua Institute of Zhejiang University, Jinhua, Zhejiang, 321299, China.
| |
Collapse
|
23
|
Dixit H, Upadhyay V, Kulharia M, Verma SK. The Study of Metalloproteome of DNA Viruses: Identification, Functional Annotation, and Diversity Analysis of Viral Metal-Binding Proteins. J Proteome Res 2024; 23:4014-4026. [PMID: 39134029 DOI: 10.1021/acs.jproteome.4c00358] [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: 09/07/2024]
Abstract
Metalloproteins are fundamental to diverse biological processes but still lack extensive investigation in viral contexts. This study reveals the prevalence and functional diversity of metal-binding proteins in DNA viruses. Among a subset of 1432 metalloproteins, zinc and magnesium-binding proteins are notably abundant, indicating their importance in viral biology. Furthermore, significant numbers of proteins binding to iron, manganese, copper, nickel, mercury, and cadmium were also detected. Human-infecting viral proteins displayed a rich landscape of metalloproteins, with MeBiPred (964 proteins) and Pfam (666) yielding the highest numbers. Interestingly, many essential viral proteins exhibited metal-binding capabilities, including polymerases, DNA binding proteins, helicases, dUPTase, thymidine kinase, and various structural and accessory proteins. This study sheds light on the ubiquitous presence of metalloproteins, their functional signatures, subcellular placements, and metal-utilization patterns, providing valuable insights into viral biology. A similar metal utilization pattern was observed in similar functional proteins across the various DNA viruses. Furthermore, these findings provide a foundation for identifying potential drug targets for combating viral infections.
Collapse
Affiliation(s)
- Himisha Dixit
- Centre for Computational Biology & Bioinformatics, Central University of Himachal Pradesh, Kangra, 176206, Himachal Pradesh, India
| | - Vipin Upadhyay
- Centre for Computational Biology & Bioinformatics, Central University of Himachal Pradesh, Kangra, 176206, Himachal Pradesh, India
| | - Mahesh Kulharia
- Centre for Computational Biology & Bioinformatics, Central University of Himachal Pradesh, Kangra, 176206, Himachal Pradesh, India
| | | |
Collapse
|
24
|
Wei M, Han C, Zhou X, Tong T, Zhang J, Ji X, Zhang P, Zhang Y, Liu Y, Zhang X, Cai T, Xie C. Filamentous morphology engineering of bacteria by iron metabolism modulation through MagR expression. Synth Syst Biotechnol 2024; 9:522-530. [PMID: 38645975 PMCID: PMC11031723 DOI: 10.1016/j.synbio.2024.04.009] [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: 01/14/2024] [Revised: 03/16/2024] [Accepted: 04/08/2024] [Indexed: 04/23/2024] Open
Abstract
The morphology is the consequence of evolution and adaptation. Escherichia coli is rod-shaped bacillus with regular dimension of about 1.5 μm long and 0.5 μm wide. Many shape-related genes have been identified and used in morphology engineering of this bacteria. However, little is known about if specific metabolism and metal irons could modulate bacteria morphology. Here in this study, we discovered filamentous shape change of E. coli cells overexpressing pigeon MagR, a putative magnetoreceptor and extremely conserved iron-sulfur protein. Comparative transcriptomic analysis strongly suggested that the iron metabolism change and iron accumulation due to the overproduction of MagR was the key to the morphological change. This model was further validated, and filamentous morphological change was also achieved by supplement E. coli cells with iron in culture medium or by increase the iron uptake genes such as entB and fepA. Our study extended our understanding of morphology regulation of bacteria, and may also serves as a prototype of morphology engineering by modulating the iron metabolism.
Collapse
Affiliation(s)
- Mengke Wei
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230039, China
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Science Island, Hefei, Anhui, 230031, China
| | - Chenyang Han
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230039, China
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Science Island, Hefei, Anhui, 230031, China
| | - Xiujuan Zhou
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Science Island, Hefei, Anhui, 230031, China
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, Anhui, 230036, China
| | - Tianyang Tong
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Science Island, Hefei, Anhui, 230031, China
- Department of Anatomy, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Jing Zhang
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Science Island, Hefei, Anhui, 230031, China
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, Anhui, 230036, China
| | - Xinmiao Ji
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Science Island, Hefei, Anhui, 230031, China
| | - Peng Zhang
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Science Island, Hefei, Anhui, 230031, China
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, Anhui, 230036, China
| | - Yanqi Zhang
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Science Island, Hefei, Anhui, 230031, China
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, Anhui, 230036, China
| | - Yan Liu
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Science Island, Hefei, Anhui, 230031, China
- Department of Anatomy, Anhui Medical University, Hefei, Anhui, 230032, China
| | - Xin Zhang
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230039, China
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Science Island, Hefei, Anhui, 230031, China
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, Anhui, 230036, China
| | - Tiantian Cai
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Science Island, Hefei, Anhui, 230031, China
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, Anhui, 230036, China
- Institute of Quantum Sensing, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| | - Can Xie
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Science Island, Hefei, Anhui, 230031, China
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, Anhui, 230036, China
- Institute of Quantum Sensing, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| |
Collapse
|
25
|
Mpakali A, Georgaki G, Buson A, Findlay AD, Foot JS, Mauvais F, van Endert P, Giastas P, Hamprecht DW, Stratikos E. Stabilization of the open conformation οf insulin-regulated aminopeptidase by a novel substrate-selective small-molecule inhibitor. Protein Sci 2024; 33:e5151. [PMID: 39167040 PMCID: PMC11337929 DOI: 10.1002/pro.5151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/30/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024]
Abstract
Insulin-regulated aminopeptidase (IRAP) is an enzyme with important biological functions and the target of drug-discovery efforts. We combined in silico screening with a medicinal chemistry optimization campaign to discover a nanomolar inhibitor of IRAP based on a pyrazolylpyrimidine scaffold. This compound displays an excellent selectivity profile versus homologous aminopeptidases, and kinetic analysis suggests it utilizes an uncompetitive mechanism of action when inhibiting the cleavage of a typical dipeptidic substrate. Surprisingly, the compound is a poor inhibitor of the processing of the physiological cyclic peptide substrate oxytocin and a 10mer antigenic epitope precursor but displays a biphasic inhibition profile for the trimming of a 9mer antigenic peptide. While the compound reduces IRAP-dependent cross-presentation of an 8mer epitope in a cellular assay, it fails to block in vitro trimming of select epitope precursors. To gain insight into the mechanism and basis of this unusual selectivity for this inhibitor, we solved the crystal structure of its complex with IRAP. The structure indicated direct zinc(II) engagement by the pyrazolylpyrimidine scaffold and revealed that the compound binds to an open conformation of the enzyme in a pose that should block the conformational transition to the enzymatically active closed conformation previously observed for other low-molecular-weight inhibitors. This compound constitutes the first IRAP inhibitor targeting the active site that utilizes a conformation-specific mechanism of action, provides insight into the intricacies of the IRAP catalytic cycle, and highlights a novel approach to regulating IRAP activity by blocking its conformational rearrangements.
Collapse
Affiliation(s)
- Anastasia Mpakali
- National Centre for Scientific Research DemokritosAthensGreece
- Laboratory of Biochemistry, Department of ChemistryNational and Kapodistrian University of AthensAthensGreece
| | - Galateia Georgaki
- National Centre for Scientific Research DemokritosAthensGreece
- Laboratory of Biochemistry, Department of ChemistryNational and Kapodistrian University of AthensAthensGreece
| | | | | | | | | | - Peter van Endert
- INSERM, CNRS, Institut Necker Enfants MaladesUniversité Paris CitéParisFrance
| | - Petros Giastas
- Department of Biotechnology, School of Applied Biology & BiotechnologyAgricultural University of AthensAthensGreece
| | | | - Efstratios Stratikos
- National Centre for Scientific Research DemokritosAthensGreece
- Laboratory of Biochemistry, Department of ChemistryNational and Kapodistrian University of AthensAthensGreece
| |
Collapse
|
26
|
Fatima N, Khalid S, Rasool N, Imran M, Parveen B, Kanwal A, Irimie M, Ciurea CI. Approachable Synthetic Methodologies for Second-Generation β-Lactamase Inhibitors: A Review. Pharmaceuticals (Basel) 2024; 17:1108. [PMID: 39338273 PMCID: PMC11434895 DOI: 10.3390/ph17091108] [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: 06/25/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
Some antibiotics that are frequently employed are β-lactams. In light of the hydrolytic process of β-lactamase, found in Gram-negative bacteria, inhibitors of β-lactamase (BLIs) have been produced. Examples of first-generation β-lactamase inhibitors include sulbactam, clavulanic acid, and tazobactam. Many kinds of bacteria immune to inhibitors have appeared, and none cover all the β-lactamase classes. Various methods have been utilized to develop second-generation β-lactamase inhibitors possessing new structures and facilitate the formation of diazabicyclooctane (DBO), cyclic boronate, metallo-, and dual-nature β-lactamase inhibitors. This review describes numerous promising second-generation β-lactamase inhibitors, including vaborbactam, avibactam, and cyclic boronate serine-β-lactamase inhibitors. Furthermore, it covers developments and methods for synthesizing MβL (metallo-β-lactamase inhibitors), which are clinically effective, as well as the various dual-nature-based inhibitors of β-lactamases that have been developed. Several combinations are still only used in preclinical or clinical research, although only a few are currently used in clinics. This review comprises materials on the research progress of BLIs over the last five years. It highlights the ongoing need to produce new and unique BLIs to counter the appearance of multidrug-resistant bacteria. At present, second-generation BLIs represent an efficient and successful strategy.
Collapse
Affiliation(s)
- Noor Fatima
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Shehla Khalid
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Nasir Rasool
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Muhammad Imran
- Chemistry Department, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
| | - Bushra Parveen
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Aqsa Kanwal
- Department of Chemistry, Government College University, Faisalabad 38000, Pakistan
| | - Marius Irimie
- Faculty of Medicine, Transylvania University of Brasov, 500036 Brasov, Romania
| | - Codrut Ioan Ciurea
- Faculty of Medicine, Transylvania University of Brasov, 500036 Brasov, Romania
| |
Collapse
|
27
|
Sawetaji, Aggarwal KK. A Protease from Moringa oleifera Lam. Exhibits In-vitro Blood Clot Solubilization and Fibrin Hydrolysis. Protein J 2024; 43:923-934. [PMID: 39068632 DOI: 10.1007/s10930-024-10222-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 07/30/2024]
Abstract
Thrombosis is the formation of abnormal blood clots in the blood vessels that obstruct blood flow and lead to thrombosis. Current treatments for thrombosis are associated with serious side effects. Therefore there is a need for alternative natural therapy. A fibrinolytic protease was isolated from fresh leaves of Moringa oleifera Lam. and characterized for its potential to solubilize blood clots and hydrolyse fibrin under in-vitro conditions. The isolated protease showed a single protein band on native-PAGE. It showed optimum fibrinolytic activity at pH 8.0, 37 oC with 50 µg protein. The fibrinolytic activity of isolated protease was also confirmed by fibrin zymography. Km and Vmax of isolated protease were determined by the Lineweaver Burk plot. The isolated protease could solubilize 96.41% of blood clots by 96 h under in-vitro conditions. In-vitro fibrin hydrolysis and blood clot solubilization activities shown by an isolated protease from leaves of Moringa oleifera Lam. suggest its fibrinolytic potential to dissolve blood clots. Being a natural molecule and from a dietary plant it can be explored as an alternative natural therapy against thrombosis.
Collapse
Affiliation(s)
- Sawetaji
- University School of Biotechnology, Guru Gobind Singh Indraprastha University, Sector 16-C Dwarka, New Delhi, 110078, India
| | - Kamal Krishan Aggarwal
- University School of Biotechnology, Guru Gobind Singh Indraprastha University, Sector 16-C Dwarka, New Delhi, 110078, India.
| |
Collapse
|
28
|
Price S, Que EL. Probing metalloenzyme dynamics in living systems: Contemporary advances in fluorescence imaging tools and applications. Curr Opin Chem Biol 2024; 81:102475. [PMID: 38852500 PMCID: PMC11715550 DOI: 10.1016/j.cbpa.2024.102475] [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: 02/20/2024] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 06/11/2024]
Abstract
Metalloenzymes are essential to cellular function, and their overexpression or enhanced activation are potential therapeutic targets. However, the study of metalloenzymes in vitro presents various challenges, leading many to develop tools to study them in their native cellular environment. Small-molecule fluorescence probes are commonly used to monitor metalloenzyme function, activity, and distribution in situ. These include probes that are activity-based (fluorescence is mediated by enzyme activity) or binding-based (fluorescence is mediated by interactions with the enzyme upon binding its metal cofactor). We discuss recent innovations that overcome key design challenges, such as the rapid diffusion of activity-based probes, the difficulty of probing redox-active enzymes, the selectivity of binding-based probes, and the poor penetration depth of fluorescence, and describe novel applications of these tools.
Collapse
Affiliation(s)
- Sky Price
- Department of Chemistry, The University of Texas at Austin, Austin, TX 78712, USA
| | - Emily L Que
- Department of Chemistry, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
29
|
Sanders MA, Chittari SS, Foley JR, Swofford WM, Elder BM, Knight AS. Leveraging Triphenylphosphine-Containing Polymers to Explore Design Principles for Protein-Mimetic Catalysts. J Am Chem Soc 2024; 146:17404-17413. [PMID: 38863219 DOI: 10.1021/jacs.4c05040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2024]
Abstract
Complex interactions between noncoordinating residues are significant yet commonly overlooked components of macromolecular catalyst function. While these interactions have been demonstrated to impact binding affinities and catalytic rates in metalloenzymes, the roles of similar structural elements in synthetic polymeric catalysts remain underexplored. Using a model Suzuki-Miyuara cross-coupling reaction, we performed a series of systematic studies to probe the interconnected effects of metal-ligand cross-links, electrostatic interactions, and local rigidity in polymer catalysts. To achieve this, a novel bifunctional triphenylphosphine acrylamide (BisTPPAm) monomer was synthesized and evaluated alongside an analogous monofunctional triphenylphosphine acrylamide (TPPAm). In model copolymer catalysts, increased initial reaction rates were observed for copolymers untethered by Pd complexation (BisTPPAm-containing) compared to Pd-cross-linked catalysts (TPPAm-containing). Further, incorporating local rigidity through secondary structure-like and electrostatic interactions revealed nonmonotonic relationships between composition and the reaction rate, demonstrating the potential for tunable behavior through secondary-sphere interactions. Finally, through rigorous cheminformatics featurization strategies and statistical modeling, we quantitated relationships between chemical descriptors of the substrate and reaction conditions on catalytic performance. Collectively, these results provide insights into relationships among the composition, structure, and function of protein-mimetic catalytic copolymers.
Collapse
Affiliation(s)
- Matthew A Sanders
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Supraja S Chittari
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Jack R Foley
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - William M Swofford
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Bridgette M Elder
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Abigail S Knight
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
30
|
Tripathi A, Dubey KD. The mechanistic insights into different aspects of promiscuity in metalloenzymes. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 141:23-66. [PMID: 38960476 DOI: 10.1016/bs.apcsb.2023.12.022] [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: 07/05/2024]
Abstract
Enzymes are nature's ultimate machinery to catalyze complex reactions. Though enzymes are evolved to catalyze specific reactions, they also show significant promiscuity in reactions and substrate selection. Metalloenzymes contain a metal ion or metal cofactor in their active site, which is crucial in their catalytic activity. Depending on the metal and its coordination environment, the metal ion or cofactor may function as a Lewis acid or base and a redox center and thus can catalyze a plethora of natural reactions. In fact, the versatility in the oxidation state of the metal ions provides metalloenzymes with a high level of catalytic adaptability and promiscuity. In this chapter, we discuss different aspects of promiscuity in metalloenzymes by using several recent experimental and theoretical works as case studies. We start our discussion by introducing the concept of promiscuity and then we delve into the mechanistic insight into promiscuity at the molecular level.
Collapse
Affiliation(s)
- Ankita Tripathi
- Department of Chemistry, School of Natural Science, Shiv Nadar Institution of Eminence, Greater Noida, Uttar Pradesh, India
| | - Kshatresh Dutta Dubey
- Department of Chemistry, School of Natural Science, Shiv Nadar Institution of Eminence, Greater Noida, Uttar Pradesh, India.
| |
Collapse
|
31
|
Miglioli F, Joel S, Tegoni M, Neira-Pelén P, Günther S, Carcelli M, Fisicaro E, Brancale A, Fernández-García Y, Rogolino D. Inhibitory interactions of the 2,3-dihydro-6,7-dihydroxy-1H-isoindol-1-one scaffold with Bunyavirales cap-snatching endonucleases expose relevant drug design features. Eur J Med Chem 2024; 272:116467. [PMID: 38735150 DOI: 10.1016/j.ejmech.2024.116467] [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: 03/04/2024] [Revised: 04/19/2024] [Accepted: 04/28/2024] [Indexed: 05/14/2024]
Abstract
The World Health Organization (WHO) identifies several bunyaviruses as significant threats to global public health security. Developing effective therapies against these viruses is crucial to combat future outbreaks and mitigate their impact on patient outcomes. Here, we report the synthesis of some isoindol-1-one derivatives and explore their inhibitory properties over an indispensable metal-dependent cap-snatching endonuclease (Cap-ENDO) shared among evolutionary divergent bunyaviruses. The compounds suppressed RNA hydrolysis by Cap-ENDOs, with IC50 values predominantly in the lower μM range. Molecular docking studies revealed the interactions with metal ions to be essential for the 2,3-dihydro-6,7-dihydroxy-1H-isoindol-1-one scaffold activity. Calorimetric analysis uncovered Mn2+ ions to have the highest affinity for sites within the targets, irrespective of aminoacidic variations influencing metal cofactor preferences. Interestingly, spectrophotometric findings unveiled sole dinuclear species formation between the scaffold and Mn2+. Moreover, the complexation of two Mn2+ ions within the viral enzymes appears to be favourable, as indicated by the binding of compound 11 to TOSV Cap-ENDO (Kd = 28 ± 3 μM). Additionally, the tendency of compound 11 to stabilize His+ more than His- Cap-ENDOs suggests exploitable differences in their catalytic pockets relevant to improving specificity. Collectively, our results underscore the isoindolinone scaffold's potential as a strategic starting point for the design of pan-antibunyavirus drugs.
Collapse
Affiliation(s)
- Francesca Miglioli
- Department of Chemistry, Life Sciences, Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124, Parma, Italy
| | - Shindhuja Joel
- Department of Virology, Bernhard-Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Matteo Tegoni
- Department of Chemistry, Life Sciences, Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124, Parma, Italy
| | - Pedro Neira-Pelén
- Department of Virology, Bernhard-Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Stephan Günther
- Department of Virology, Bernhard-Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Mauro Carcelli
- Department of Chemistry, Life Sciences, Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124, Parma, Italy
| | - Emilia Fisicaro
- Department of Food and Drug, University of Parma, Viale delle Scienze 27/A, 43124, Parma, Italy
| | - Andrea Brancale
- Department of Organic Chemistry, University of Chemistry and Technology, 16628, Prague, Czech Republic
| | - Yaiza Fernández-García
- Department of Virology, Bernhard-Nocht Institute for Tropical Medicine, Hamburg, Germany.
| | - Dominga Rogolino
- Department of Chemistry, Life Sciences, Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/A, 43124, Parma, Italy.
| |
Collapse
|
32
|
Kovács H, Jakusch T, May NV, Tóth S, Szakács G, Enyedy ÉA. Complex formation of ML324, the histone demethylase inhibitor, with essential metal ions: Relationship between solution chemistry and anticancer activity. J Inorg Biochem 2024; 255:112540. [PMID: 38552361 DOI: 10.1016/j.jinorgbio.2024.112540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/15/2024] [Accepted: 03/22/2024] [Indexed: 04/16/2024]
Abstract
N-(3-(dimethylamino)propyl-4-(8-hydroxyquinolin-6-yl)benzamide (ML324, HL) is a potent inhibitor of the iron-containing histone demethylase KDM4, a recognized potential target of cancer therapeutics. Herein, we report the proton dissociation and complex formation processes of ML324 with essential metal ions such as Fe(II), Fe(III), Cu(II) and Zn(II) using UV-visible, fluorescence, electron paramagnetic resonance and 1H NMR spectroscopic methods. The electrochemical behaviour of the copper and iron complexes was characterized by cyclic voltammetry and spectroelectrochemistry. The solid phase structure of ML324 analysed by X-ray crystallography is also provided. Based on the solution equilibrium data, ML324 is present in solution in H2L+ form with a protonated dimethylammonium moiety at pH 7.4, and this (N,O) donor bearing ligand forms mono and bis complexes with all the studied metal ions and the tris-ligand species is also observed with Fe(III). At pH 7.4 the metal binding ability of ML324 follows the order: Fe(II) < Zn(II) < Cu(II) < Fe(III). Complexation with iron resulted in a negative redox potential (E'1/2 = -145 mV vs. NHE), further suggesting that the ligand has a preference for Fe(III) over Fe(II). ML324 was tested for its anticancer activity in chemosensitive and resistant human cancer cells overexpressing the efflux pump P-glycoprotein. ML324 exerted similar activity in all tested cells (IC50 = 1.9-3.6 μM). Co-incubation and complexation of the compound with Cu(II) and Zn(II) had no impact on the cytotoxicity of ML324, whereas Fe(III) decreased the toxicity in a concentration-dependent manner, and this effect was more pronounced in the multidrug resistant cells.
Collapse
Affiliation(s)
- Hilda Kovács
- MTA-SZTE Lendület Functional Metal Complexes Research Group, Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8, H-6720 Szeged, Hungary; Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8, H-6720 Szeged, Hungary
| | - Tamás Jakusch
- Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8, H-6720 Szeged, Hungary
| | - Nóra V May
- Centre for Structural Science, Research Centre for Natural Sciences, Hungarian Research Network (HUN-REN), Magyar tudósok körútja 2, H-1117 Budapest, Hungary
| | - Szilárd Tóth
- Drug Resistance Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, Hungarian Research Network (HUN-REN), Magyar Tudósok krt. 2, H-1117 Budapest, Hungary; National Laboratory for Drug Research and Development, Magyar Tudósok krt. 2, H-1117 Budapest, Hungary
| | - Gergely Szakács
- Drug Resistance Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, Hungarian Research Network (HUN-REN), Magyar Tudósok krt. 2, H-1117 Budapest, Hungary; Center for Cancer Research, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | - Éva A Enyedy
- MTA-SZTE Lendület Functional Metal Complexes Research Group, Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8, H-6720 Szeged, Hungary; Department of Molecular and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7-8, H-6720 Szeged, Hungary.
| |
Collapse
|
33
|
Li H, Pang J, Hu W, Caballero V, Sun J, Tan M, Hu JZ, Ni Y, Wang Y. Confined dual Lewis acid centers for selective cascade C-C coupling and deoxygenation. Chem Sci 2024; 15:8031-8037. [PMID: 38817567 PMCID: PMC11134334 DOI: 10.1039/d3sc06921d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 04/11/2024] [Indexed: 06/01/2024] Open
Abstract
The selective formation of C-C bonds, coupled with effective removal of oxygen, plays a crucial role in the process of upgrading biomass-derived oxygenates into fuels and chemicals. However, co-feeding reactants with water is sometimes necessary to assist binding sites in catalytic reactions, thereby achieving desirable performance. Here, we report the design of a CeSnBeta catalyst featuring dual Lewis acidic sites for the efficient production of isobutene from acetone via C-C coupling followed by deoxygenation. By incorporating Ce species onto SnBeta, which was synthesized through liquid-phase grafting of dealuminated Beta, we created confined dual Lewis acidic centers within Beta zeolites. The cooperative action of Ce species and framework Sn sites within this confined environment enabled selective catalysis of the acetone-to-isobutene cascade reactions, showcasing enhanced stability even without the presence of water.
Collapse
Affiliation(s)
- Houqian Li
- The Gene & Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University Pullman WA 99164 USA
| | - Jifeng Pang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences No. 457 Zhongshan Road Dalian 116023 P.R. China
| | - Wenda Hu
- The Gene & Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University Pullman WA 99164 USA
- Pacific Northwest National Laboratory Richland WA 99352 USA
| | - Vannessa Caballero
- The Gene & Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University Pullman WA 99164 USA
| | - Junming Sun
- The Gene & Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University Pullman WA 99164 USA
| | - Mingwu Tan
- Institute of Sustainability for Chemicals, Energy and Environment 1 Pesek Road Jurong Island 627833 Singapore
| | - Jian Zhi Hu
- Pacific Northwest National Laboratory Richland WA 99352 USA
| | - Yelin Ni
- Pacific Northwest National Laboratory Richland WA 99352 USA
| | - Yong Wang
- The Gene & Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University Pullman WA 99164 USA
- Pacific Northwest National Laboratory Richland WA 99352 USA
| |
Collapse
|
34
|
Rossi S, Tudino V, Carullo G, Butini S, Campiani G, Gemma S. Metalloenzyme Inhibitors against Zoonotic Infections: Focus on Leishmania and Schistosoma. ACS Infect Dis 2024; 10:1520-1535. [PMID: 38669567 DOI: 10.1021/acsinfecdis.4c00163] [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: 04/28/2024]
Abstract
The term "zoonosis" denotes diseases transmissible among vertebrate animals and humans. These diseases constitute a significant public health challenge, comprising 61% of human pathogens and causing an estimated 2.7 million deaths annually. Zoonoses not only affect human health but also impact animal welfare and economic stability, particularly in low- and middle-income nations. Leishmaniasis and schistosomiasis are two important neglected tropical diseases with a high prevalence in tropical and subtropical areas, imposing significant burdens on affected regions. Schistosomiasis, particularly rampant in sub-Saharan Africa, lacks alternative treatments to praziquantel, prompting concerns regarding parasite resistance. Similarly, leishmaniasis poses challenges with unsatisfactory treatments, urging the development of novel therapeutic strategies. Effective prevention demands a One Health approach, integrating diverse disciplines to enhance diagnostics and develop safer drugs. Metalloenzymes, involved in parasite biology and critical in different biological pathways, emerged in the last few years as useful drug targets for the treatment of human diseases. Herein we have reviewed recent reports on the discovery of inhibitors of metalloenzymes associated with zoonotic diseases like histone deacetylases (HDACs), carbonic anhydrase (CA), arginase, and heme-dependent enzymes.
Collapse
Affiliation(s)
- Sara Rossi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Valeria Tudino
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Gabriele Carullo
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
- Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan 81746-7346, Iran
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| |
Collapse
|
35
|
Karthikeyan D, Kumar S, Jayaprakash NS. A comprehensive review of recent developments in the gram-negative bacterial UDP-2,3-diacylglucosamine hydrolase (LpxH) enzyme. Int J Biol Macromol 2024; 267:131327. [PMID: 38574903 DOI: 10.1016/j.ijbiomac.2024.131327] [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: 11/20/2023] [Revised: 03/09/2024] [Accepted: 03/31/2024] [Indexed: 04/06/2024]
Abstract
The emergence of multidrug resistance has provided a great challenge to treat nosocomial infections, which have become a major health threat around the globe. Lipid A (an active endotoxin component), the final product of the Raetz lipid A metabolism pathway, is a membrane anchor of lipopolysaccharide (LPS) of the gram-negative bacterial outer membrane. It shields bacterial cells and serves as a protective barrier from antibiotics, thereby eliciting host response and making it difficult to destroy. UDP-2,3-diacylglucosamine pyrophosphate hydrolase (LpxH), a crucial peripheral membrane enzyme of the Raetz pathway, turned out to be the potential target to inhibit the production of Lipid A. This review provides a comprehensive compilation of information regarding the structural and functional aspects of LpxH, as well as its analogous LpxI and LpxG. In addition, apart from by providing a broader understanding of the enzyme-inhibitor mechanism, this review facilitates the development of novel drug candidates that can inhibit the pathogenicity of the lethal bacterium.
Collapse
Affiliation(s)
- Divyapriya Karthikeyan
- Centre for Bioseparation Technology, Vellore Institute of Technology, Vellore 632014, India
| | - Sanjit Kumar
- Department of Biotechnology, School of Interdisciplinary Education and Research, Guru Ghasidas Vishwavidyalaya, Bilaspur (A Central University), Chhattisgarh 495009, India
| | - N S Jayaprakash
- Centre for Bioseparation Technology, Vellore Institute of Technology, Vellore 632014, India.
| |
Collapse
|
36
|
Viana LPS, Naves GM, Medeiros IG, Guimarães AS, Sousa ES, Santos JCC, Freire NML, de Aquino TM, Modolo LV, de Fátima Â, da Silva CM. Synergizing structure and function: Cinnamoyl hydroxamic acids as potent urease inhibitors. Bioorg Chem 2024; 146:107247. [PMID: 38493635 DOI: 10.1016/j.bioorg.2024.107247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/16/2024] [Accepted: 02/27/2024] [Indexed: 03/19/2024]
Abstract
The current investigation encompasses the structural planning, synthesis, and evaluation of the urease inhibitory activity of a series of molecular hybrids of hydroxamic acids and Michael acceptors, delineated from the structure of cinnamic acids. The synthesized compounds exhibited potent urease inhibitory effects, with IC50 values ranging from 3.8 to 12.8 µM. Kinetic experiments unveiled that the majority of the synthesized hybrids display characteristics of mixed inhibitors. Generally, derivatives containing electron-withdrawing groups on the aromatic ring demonstrate heightened activity, indicating that the increased electrophilicity of the beta carbon in the Michael Acceptor moiety positively influences the antiureolytic properties of this compounds class. Biophysical and theoretical investigations further corroborated the findings obtained from kinetic assays. These studies suggest that the hydroxamic acid core interacts with the urease active site, while the Michael acceptor moiety binds to one or more allosteric sites adjacent to the active site.
Collapse
Affiliation(s)
- Luciana P S Viana
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Giovanna M Naves
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Isabela G Medeiros
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ari S Guimarães
- Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Maceió, AL, Brazil
| | - Emilly S Sousa
- Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Maceió, AL, Brazil
| | - Josué C C Santos
- Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Maceió, AL, Brazil
| | - Nathália M L Freire
- Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Maceió, AL, Brazil
| | - Thiago M de Aquino
- Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Maceió, AL, Brazil
| | - Luzia V Modolo
- Departamento de Botânica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ângelo de Fátima
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Cleiton M da Silva
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
37
|
Ermini E, Brai A, Cini E, Finetti F, Giannini G, Padula D, Paradisi L, Poggialini F, Trabalzini L, Tolu P, Taddei M. A novel bioresponsive self-immolative spacer based on aza-quinone methide reactivity for the controlled release of thiols, phenols, amines, sulfonamides or amides. Chem Sci 2024; 15:6168-6177. [PMID: 38665538 PMCID: PMC11041255 DOI: 10.1039/d4sc01576b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 03/24/2024] [Indexed: 04/28/2024] Open
Abstract
A stimuli-sensitive linker is one of the indispensable components of prodrugs for cancer therapy as it covalently binds the drug and releases it upon external stimulation at the tumour site. Quinone methide elimination has been widely used as the key transformation to release drugs based on their nucleofugacity. The usual approach is to bind the drug to the linker as a carbamate and release it as a free amine after a self-immolative 1,6-elimination. Although this approach is very efficient, it is limited to amines (as carbamates), alcohols or phenols (as carbonates) or other acidic functional groups. We report here a self-immolative spacer capable of directly linking and releasing amines, phenols, thiols, sulfonamides and carboxyamides after a reductive stimulus. The spacer is based on the structure of (5-nitro-2-pyrrolyl)methanol (NPYM-OH), which was used for the direct alkylation of the functional groups mentioned above. The spacer is metabolically stable and has three indispensable sites for bioconjugation: the bioresponsive trigger, the conjugated 1,6 self-immolative system and a third arm suitable for conjugation with a carrier or other modifiers. Release was achieved by selective reduction of the nitro group over Fe/Pd nanoparticles (NPs) in a micellar aqueous environment (H2O/TPGS-750-M), or by NADH mediated nitroreductase activation. A DFT study demonstrates that, during the 1,6 elimination, the transition state formed from 5-aminopyrrole has a lower activation energy compared to other 5-membered heterocycles or p-aminobenzyl derivatives. The NPYM scaffold was validated by late-stage functionalisation of approved drugs such as celecoxib, colchicine, vorinostat or ciprofloxacin. A hypoxia-activated NPYM-based prodrug (HAP) derived from HDAC inhibitor ST7612AA1 was also produced, which was active in cancer cells under hypoxic conditions.
Collapse
Affiliation(s)
- Elena Ermini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena Via A. Moro 2 53100 Siena Italy
| | - Annalaura Brai
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena Via A. Moro 2 53100 Siena Italy
| | - Elena Cini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena Via A. Moro 2 53100 Siena Italy
| | - Federica Finetti
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena Via A. Moro 2 53100 Siena Italy
| | - Giuseppe Giannini
- Translational Medicine & Clinical Pharmacology Corporate R&D - Alfasigma SpA Via Pontina, km 30400 00071 Pomezia (Roma) Italy
| | - Daniele Padula
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena Via A. Moro 2 53100 Siena Italy
| | - Lucrezia Paradisi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena Via A. Moro 2 53100 Siena Italy
| | - Federica Poggialini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena Via A. Moro 2 53100 Siena Italy
| | - Lorenza Trabalzini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena Via A. Moro 2 53100 Siena Italy
| | - Paola Tolu
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena Via A. Moro 2 53100 Siena Italy
| | - Maurizio Taddei
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena Via A. Moro 2 53100 Siena Italy
| |
Collapse
|
38
|
Han D, Yu F, Zheng L. Causal effect of serum matrix metalloproteinase levels on venous thromboembolism: a Mendelian randomization study. Epidemiol Health 2024; 46:e2024046. [PMID: 38697862 PMCID: PMC11417446 DOI: 10.4178/epih.e2024046] [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: 03/06/2024] [Accepted: 03/26/2024] [Indexed: 05/05/2024] Open
Abstract
OBJECTIVES Serum matrix metalloproteinase (MMP) levels are associated with cardiovascular diseases. However, the causal associations between serum levels of specific MMPs and venous thromboembolism (VTE) remain unclear. The present study sought to explore the causal relationship between serum MMP levels and VTE by using the Mendelian randomization (MR) method. METHODS In this study 2-sample MR study, the exposure data on serum MMP levels were derived from genome-wide association studies involving 21,758 individuals from 13 cohorts of European descent. The outcome data on VTE, including deep vein thrombosis and pulmonary embolism, were derived from the FinnGen research project. The primary method used was the inverse-variance weighting method. The MR-Egger intercept test and the Cochran Q test were used to evaluate pleiotropy and heterogeneity. RESULTS Using the inverse-variance weighting method, higher serum MMP-12 levels were found to be associated with an increased risk of VTE (odds ratio, 1.04; 95% confidence interval, 1.01 to 1.07; p=0.001). Moreover, there was a weak association between the levels of certain MMPs and VTE. Sensitivity analyses revealed no significant heterogeneity and pleiotropy in our study, and the Steiger directionality test did not reveal a significant reverse causation association. CONCLUSIONS There is a causal association between MMP-12 levels and VTE, which may have substantial implications for the diagnostic and therapeutic strategies used for VTE.
Collapse
Affiliation(s)
- Deheng Han
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fangcong Yu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liangrong Zheng
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
39
|
Schuck B, Brenk R. On the hunt for metalloenzyme inhibitors: Investigating the presence of metal-coordinating compounds in screening libraries and chemical spaces. Arch Pharm (Weinheim) 2024; 357:e2300648. [PMID: 38279543 DOI: 10.1002/ardp.202300648] [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: 11/08/2023] [Revised: 12/20/2023] [Accepted: 12/27/2023] [Indexed: 01/28/2024]
Abstract
Metalloenzymes play vital roles in various biological processes, requiring the search for inhibitors to develop treatment options for diverse diseases. While compound library screening is a conventional approach, the exploration of virtual chemical spaces housing trillions of compounds has emerged as an alternative strategy. In this study, we investigated the suitability of selected screening libraries and chemical spaces for discovering inhibitors of metalloenzymes featuring common ions (Mg2+, Mn2+, and Zn2+). First, metal-coordinating groups from ligands interacting with ions in the Protein Data Bank were extracted. Subsequently, the prevalence of these groups in two focused screening libraries (Life Chemicals' chelator library, comprising 6,428 compounds, and Otava's chelator fragment library, with 1,784 fragments) as well as two chemical spaces (GalaXi and REAL space, containing billions of virtual products) was investigated. In total, 1,223 metal-coordinating groups were identified, with about a quarter of these groups found within the examined libraries and spaces. Our results indicate that these can serve as valuable starting points for drug discovery targeting metalloenzymes. In addition, this study suggests ways to improve libraries and spaces for better success in finding potential inhibitors for metalloenzymes.
Collapse
Affiliation(s)
- Bruna Schuck
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Ruth Brenk
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Computational Biology Unit, University of Bergen, Bergen, Norway
| |
Collapse
|
40
|
Dawbaa S, Türkeş C, Nuha D, Demir Y, Evren AE, Yurttaş L, Beydemir Ş. New N-(1,3,4-thiadiazole-2-yl)acetamide derivatives as human carbonic anhydrase I and II and acetylcholinesterase inhibitors. J Biomol Struct Dyn 2024:1-19. [PMID: 38533902 DOI: 10.1080/07391102.2024.2331085] [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: 09/19/2023] [Accepted: 03/11/2024] [Indexed: 03/28/2024]
Abstract
Various carbonic anhydrase (CA) enzyme isoforms are known today. In addition to the use of CA inhibitors as diuretics, antiepileptics and antiglaucoma agents, the inhibition of other specific isoforms of CA was reported to have clinical benefits in cancers. In this study, two groups of 1,3,4-thiadiazole derivatives were designed and synthesized to act as human CA I and II (hCA I and hCA II) inhibitors. The activities of these compounds were tested in vitro and evaluated in silico studies. The activity of the synthesized compounds was also tested against acetylcholinesterase (AChE) to evaluate the relation of the newly designed structures to the activity against AChE. The synthesized compounds were analyzed by 1H NMR,13C NMR and high-resolution mass spectroscopy (HRMS). The results displayed a better activity of all the synthesized compounds against hCA I than that of the commonly used standard drug, Acetazolamide (AAZ). The compounds also showed better activity against hCA II, except for compounds 5b and 6b. Only compounds 6a and 6c showed superior activity against AChE compared to the standard agent, tacrine (THA). In silico studies, including absorption, distribution, metabolism and excretion (ADME) and drug-likeness evaluation, molecular docking, molecular dynamic simulations (MDSs) and density functional theory (DFT) calculations, were compatible with the in vitro results and presented details regarding the structure-activity relationship.
Collapse
Affiliation(s)
- Sam Dawbaa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
- Department of Doctor of Pharmacy (PharmD), Faculty of Medical Sciences, Thamar University, Dhamar, Yemen
- Department of Pharmacy, Faculty of Medical Sciences, Al-Hikma University, Dhamar, Yemen
| | - Cüneyt Türkeş
- Department of Biochemistry, Faculty of Pharmacy, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Demokrat Nuha
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
- Department of Chemistry, Faculty of Science, Eskisehir Technical University, Eskişehir, Turkey
- Faculty of Pharmacy, University for Business and Technology, Prishtina, Kosovo
| | - Yeliz Demir
- Department of Pharmacy Services, Nihat Delibalta Göle Vocational High School, Ardahan University, Ardahan, Turkey
| | - Asaf Evrim Evren
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
- Department of Pharmacy Services, Vocational School of Health Services, Bilecik Şeyh Edebali University, Bilecik, Turkey
| | - Leyla Yurttaş
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Şükrü Beydemir
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
- The Rectorate of Bilecik Seyh Edebali University, Bilecik, Turkey
| |
Collapse
|
41
|
Zhang L, Yang Y, Yang Y, Xiao Z. Discovery of Novel Metalloenzyme Inhibitors Based on Property Characterization: Strategy and Application for HDAC1 Inhibitors. Molecules 2024; 29:1096. [PMID: 38474606 DOI: 10.3390/molecules29051096] [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: 11/30/2023] [Revised: 02/17/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Metalloenzymes are ubiquitously present in the human body and are relevant to a variety of diseases. However, the development of metalloenzyme inhibitors is limited by low specificity and poor drug-likeness associated with metal-binding fragments (MBFs). A generalized drug discovery strategy was established, which is characterized by the property characterization of zinc-dependent metalloenzyme inhibitors (ZnMIs). Fifteen potential Zn2+-binding fragments (ZnBFs) were identified, and a customized pharmacophore feature was defined based on these ZnBFs. The customized feature was set as a required feature and applied to a search for novel inhibitors for histone deacetylase 1 (HDAC1). Ten potential HDAC1 inhibitors were recognized, and one of them (compound 9) was a known potent HDAC1 inhibitor. The results demonstrated the effectiveness of our strategy to identify novel inhibitors for zinc-dependent metalloenzymes.
Collapse
Affiliation(s)
- Lu Zhang
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Department of Toxicology, Tianjin Centers for Disease Control and Prevention, Tianjin 300011, China
| | - Yajun Yang
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ying Yang
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhiyan Xiao
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- State Key Laboratory of Digestive Health, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
42
|
Rayi S, Cai Y, Greenwich JL, Fuqua C, Gerdt JP. Interbacterial Biofilm Competition through a Suite of Secreted Metabolites. ACS Chem Biol 2024; 19:462-470. [PMID: 38261537 PMCID: PMC10951839 DOI: 10.1021/acschembio.3c00655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Polymicrobial biofilms are ubiquitous, and the complex interspecies interactions within them are cryptic. We discovered the chemical foundation of antagonistic interactions in a model dual-species biofilm in which Pseudomonas aeruginosa inhibits the biofilm formation of Agrobacterium tumefaciens. Three known siderophores produced by P. aeruginosa (pyoverdine, pyochelin, and dihydroaeruginoic acid) were each capable of inhibiting biofilm formation. Surprisingly, a mutant that was incapable of producing these siderophores still secreted an antibiofilm metabolite. We discovered that this inhibitor was N5-formyl-N5-hydroxy-l-ornithine (fOHOrn)─a precursor in pyoverdine biosynthesis. Unlike the siderophores, this inhibitor did not appear to function via extracellular metal sequestration. In addition to this discovery, the compensatory overproduction of a new biofilm inhibitor illustrates the risk of pleiotropy in genetic knockout experiments. In total, this work lends new insight into the chemical nature of dual-species biofilm regulation and reveals a new naturally produced inhibitor of A. tumefaciens biofilm formation.
Collapse
Affiliation(s)
- Soniya Rayi
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Yanyao Cai
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Jennifer L Greenwich
- Department of Biology, Indiana University, Bloomington, Indiana 47405, United States
| | - Clay Fuqua
- Department of Biology, Indiana University, Bloomington, Indiana 47405, United States
| | - Joseph P Gerdt
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| |
Collapse
|
43
|
Novakova Z, Tehrani ZA, Jurok R, Motlova L, Kutil Z, Pavlicek J, Shukla S, Choy CJ, Havlinova B, Baranova P, Berkman CE, Kuchar M, Cerny J, Barinka C. Structural, Biochemical, and Computational Characterization of Sulfamides as Bimetallic Peptidase Inhibitors. J Chem Inf Model 2024; 64:1030-1042. [PMID: 38224368 PMCID: PMC10865363 DOI: 10.1021/acs.jcim.3c01542] [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: 09/23/2023] [Revised: 12/19/2023] [Accepted: 12/28/2023] [Indexed: 01/16/2024]
Abstract
The sulfonamide function is used extensively as a general building block in various inhibitory scaffolds and, more specifically, as a zinc-binding group (ZBG) of metalloenzyme inhibitors. Here, we provide biochemical, structural, and computational characterization of a metallopeptidase in complex with inhibitors, where the mono- and bisubstituted sulfamide functions are designed to directly engage zinc ions of a bimetallic enzyme site. Structural data showed that while monosubstituted sulfamides coordinate active-site zinc ions via the free negatively charged amino group in a canonical manner, their bisubstituted counterparts adopt an atypical binding pattern divergent from expected positioning of corresponding tetrahedral reaction intermediates. Accompanying quantum mechanics calculations revealed that electroneutrality of the sulfamide function is a major factor contributing to the markedly lower potency of bisubstituted compounds by considerably lowering their interaction energy with the enzyme. Overall, while bisubstituted uncharged sulfamide functions can bolster favorable pharmacological properties of a given inhibitor, their use as ZBGs in metalloenzyme inhibitors might be less advantageous due to their suboptimal metal-ligand properties.
Collapse
Affiliation(s)
- Zora Novakova
- Institute
of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech
Republic
| | - Zahra Aliakbar Tehrani
- Institute
of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech
Republic
| | - Radek Jurok
- Forensic
Laboratory of Biologically Active Substances, University of Chemistry and Technology Prague, Technická 3, 166 28 Prague 6, Czech Republic
| | - Lucia Motlova
- Institute
of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech
Republic
| | - Zsofia Kutil
- Institute
of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech
Republic
| | - Jiri Pavlicek
- Institute
of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech
Republic
| | - Shivam Shukla
- Institute
of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech
Republic
| | - Cindy J. Choy
- Department
of Chemistry, Washington State University, Pullman, Washington 99163, United States
| | - Barbora Havlinova
- Institute
of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech
Republic
| | - Petra Baranova
- Institute
of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech
Republic
| | - Clifford E. Berkman
- Department
of Chemistry, Washington State University, Pullman, Washington 99163, United States
| | - Martin Kuchar
- Forensic
Laboratory of Biologically Active Substances, University of Chemistry and Technology Prague, Technická 3, 166 28 Prague 6, Czech Republic
| | - Jiri Cerny
- Institute
of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech
Republic
| | - Cyril Barinka
- Institute
of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 252 50 Vestec, Czech
Republic
| |
Collapse
|
44
|
Ibrahim IH. Metalloproteins and metalloproteomics in health and disease. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 141:123-176. [PMID: 38960472 DOI: 10.1016/bs.apcsb.2023.12.013] [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: 07/05/2024]
Abstract
Metalloproteins represents more than one third of human proteome, with huge variation in physiological functions and pathological implications, depending on the metal/metals involved and tissue context. Their functions range from catalysis, bioenergetics, redox, to DNA repair, cell proliferation, signaling, transport of vital elements, and immunity. The human metalloproteomic studies revealed that many families of metalloproteins along with individual metalloproteins are dysregulated under several clinical conditions. Also, several sorts of interaction between redox- active or redox- inert metalloproteins are observed in health and disease. Metalloproteins profiling shows distinct alterations in neurodegenerative diseases, cancer, inflammation, infection, diabetes mellitus, among other diseases. This makes metalloproteins -either individually or as families- a promising target for several therapeutic approaches. Inhibitors and activators of metalloenzymes, metal chelators, along with artificial metalloproteins could be versatile in diagnosis and treatment of several diseases, in addition to other biomedical and industrial applications.
Collapse
Affiliation(s)
- Iman Hassan Ibrahim
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt.
| |
Collapse
|
45
|
Abstract
Metalloenzymes are responsible for numerous physiological and pathological processes in living organisms; however, there are very few FDA-approved metalloenzyme-targeting therapeutics (only ~ 67 FDA-approved metalloenzyme inhibitors as of 2020, less than ~ 5 % of all FDA-approved therapeutics). Most metalloenzyme inhibitors have been developed to target the catalytic metal centers in metalloenzymes via the incorporation of metal-binding groups. Light-controlled inhibition of metalloenzymes has been used as a means to specifically activate and inactivate inhibitor engagement at a desired location and time via light irradiation, allowing for precise spatiotemporal control over metalloenzyme activity. In this review, we summarize the strategies that have been employed to develop biocompatible light-sensitive inhibitors for metalloenzymes via the incorporation of different photo-activatable moieties (including photoswitchable and photocleavable groups), and the application of photo-activateable inhibitors both in vitro and in vivo. We also discuss the photophysical mechanisms of different photo-activatable groups, their action under physiological conditions, and the different modes of interaction between inhibitors and proteins (i.e., inhibition mechanisms) in the presence and absence of light. Finally, we discuss considerations for the future development of light-responsive metalloenzyme inhibitors and the challenges limiting their application in vivo.
Collapse
Affiliation(s)
- Noushaba Nusrat Mafy
- Department of Chemistry, University of Texas at Austin, 105 E 24th St, Austin, TX 78712, United States
| | - Dorothea B. Hudson
- Department of Chemistry, University of Texas at Austin, 105 E 24th St, Austin, TX 78712, United States
| | - Emily L. Que
- Department of Chemistry, University of Texas at Austin, 105 E 24th St, Austin, TX 78712, United States
| |
Collapse
|
46
|
Xu W, Wu Y, Gu W, Du D, Lin Y, Zhu C. Atomic-level design of metalloenzyme-like active pockets in metal-organic frameworks for bioinspired catalysis. Chem Soc Rev 2024; 53:137-162. [PMID: 38018371 DOI: 10.1039/d3cs00767g] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Natural metalloenzymes with astonishing reaction activity and specificity underpin essential life transformations. Nevertheless, enzymes only operate under mild conditions to keep sophisticated structures active, limiting their potential applications. Artificial metalloenzymes that recapitulate the catalytic activity of enzymes can not only circumvent the enzymatic fragility but also bring versatile functions into practice. Among them, metal-organic frameworks (MOFs) featuring diverse and site-isolated metal sites and supramolecular structures have emerged as promising candidates for metalloenzymes to move toward unparalleled properties and behaviour of enzymes. In this review, we systematically summarize the significant advances in MOF-based metalloenzyme mimics with a special emphasis on active pocket engineering at the atomic level, including primary catalytic sites and secondary coordination spheres. Then, the deep understanding of catalytic mechanisms and their advanced applications are discussed. Finally, a perspective on this emerging frontier research is provided to advance bioinspired catalysis.
Collapse
Affiliation(s)
- Weiqing Xu
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. China.
| | - Yu Wu
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. China.
| | - Wenling Gu
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. China.
| | - Dan Du
- School of Mechanical and Materials Engineering, Washington State University, 99164, Pullman, USA.
| | - Yuehe Lin
- School of Mechanical and Materials Engineering, Washington State University, 99164, Pullman, USA.
| | - Chengzhou Zhu
- National Key Laboratory of Green Pesticide, International Joint Research Center for Intelligent Biosensing Technology and Health, College of Chemistry, Central China Normal University, Wuhan 430079, P. R. China.
| |
Collapse
|
47
|
Podili R, Mishra KMA, Akkewar AS, Kumar S, Rayala VVSPK, Kulhari U, Sahu BD, P R, Sethi KK. Design, synthesis, and histone deacetylase inhibition study of novel 4-(2-aminoethyl) phenol derivatives. J Biochem Mol Toxicol 2024; 38:e23591. [PMID: 38037273 DOI: 10.1002/jbt.23591] [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: 09/27/2023] [Revised: 11/08/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023]
Abstract
Histone deacetylases (HDACs) have been identified as promising targets for anticancer treatment. The study demonstrates virtual screening, molecular docking, and synthesis of 4-(2-aminoethyl) phenol derivatives as HDAC inhibitors. The virtual screening and molecular docking analysis led to the identification of 10 representative compounds, which were evaluated based on their drug-like properties. The results demonstrated that these compounds effectively interacted with the active site pocket of HDAC 3 through π-stacking, Zn2+ coordination, hydrogen bonding, and hydrophobic interactions with catalytic residues. Furthermore, a series of 4-(2-aminoethyl) phenol derivatives were synthesized, and their HDAC inhibitory activity was evaluated. Compounds 18 and 20 showed significant HDAC inhibitory activity of 64.94 ± 1.17% and 52.45 ± 1.45%, respectively, compared to the solvent control. The promising results of this study encourage further research on 4-(2-aminoethyl) phenol derivatives and may provide significant insight into the design of novel small molecule HDAC inhibitors to fight against target-specific malignancies of chronic obstructive pulmonary disease and nonsmall cell lung cancer in the future.
Collapse
Affiliation(s)
- Runesh Podili
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - K M Abha Mishra
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Ashish S Akkewar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Sanjay Kumar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - V V S Prasanna Kumari Rayala
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Uttam Kulhari
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Bidya D Sahu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Radhakrishnanand P
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Kalyan K Sethi
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| |
Collapse
|
48
|
Plotniece A, Sobolev A, Supuran CT, Carta F, Björkling F, Franzyk H, Yli-Kauhaluoma J, Augustyns K, Cos P, De Vooght L, Govaerts M, Aizawa J, Tammela P, Žalubovskis R. Selected strategies to fight pathogenic bacteria. J Enzyme Inhib Med Chem 2023; 38:2155816. [PMID: 36629427 PMCID: PMC9848314 DOI: 10.1080/14756366.2022.2155816] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/02/2022] [Accepted: 12/02/2022] [Indexed: 01/12/2023] Open
Abstract
Natural products and analogues are a source of antibacterial drug discovery. Considering drug resistance levels emerging for antibiotics, identification of bacterial metalloenzymes and the synthesis of selective inhibitors are interesting for antibacterial agent development. Peptide nucleic acids are attractive antisense and antigene agents representing a novel strategy to target pathogens due to their unique mechanism of action. Antisense inhibition and development of antisense peptide nucleic acids is a new approach to antibacterial agents. Due to the increased resistance of biofilms to antibiotics, alternative therapeutic options are necessary. To develop antimicrobial strategies, optimised in vitro and in vivo models are needed. In vivo models to study biofilm-related respiratory infections, device-related infections: ventilator-associated pneumonia, tissue-related infections: chronic infection models based on alginate or agar beads, methods to battle biofilm-related infections are discussed. Drug delivery in case of antibacterials often is a serious issue therefore this review includes overview of drug delivery nanosystems.
Collapse
Affiliation(s)
- Aiva Plotniece
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Riga Stradiņš University, Riga, Latvia
| | | | - Claudiu T. Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Firenze, Italy
| | - Fabrizio Carta
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Firenze, Italy
| | - Fredrik Björkling
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, Center for Peptide-Based Antibiotics, University of Copenhagen, Copenhagen East, Denmark
| | - Henrik Franzyk
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, Center for Peptide-Based Antibiotics, University of Copenhagen, Copenhagen East, Denmark
| | - Jari Yli-Kauhaluoma
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, Drug Research Program, University of Helsinki, Helsinki, Finland
| | - Koen Augustyns
- Infla-Med, Centre of Excellence, University of Antwerp, Antwerp, Belgium
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Paul Cos
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Linda De Vooght
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Matthias Govaerts
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Juliana Aizawa
- Department of Pharmaceutical Sciences, Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Antwerp, Belgium
| | - Päivi Tammela
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, Drug Research Program, University of Helsinki, Helsinki, Finland
| | - Raivis Žalubovskis
- Latvian Institute of Organic Synthesis, Riga, Latvia
- Faculty of Materials Science and Applied Chemistry, Institute of Technology of Organic Chemistry, Riga Technical University, Riga, Latvia
| |
Collapse
|
49
|
Proj M, Hrast M, Bajc G, Frlan R, Meden A, Butala M, Gobec S. Discovery of a fragment hit compound targeting D-Ala:D-Ala ligase of bacterial peptidoglycan biosynthesis. J Enzyme Inhib Med Chem 2023; 38:387-397. [PMID: 36446617 PMCID: PMC9718554 DOI: 10.1080/14756366.2022.2149745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Bacterial resistance is an increasing threat to healthcare systems, highlighting the need for discovering new antibacterial agents. An established technique, fragment-based drug discovery, was used to target a bacterial enzyme Ddl involved in the biosynthesis of peptidoglycan. We assembled general and focused fragment libraries that were screened in a biochemical inhibition assay. Screening revealed a new fragment-hit inhibitor of DdlB with a Ki value of 20.7 ± 4.5 µM. Binding to the enzyme was confirmed by an orthogonal biophysical method, surface plasmon resonance, making the hit a promising starting point for fragment development.
Collapse
Affiliation(s)
- Matic Proj
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Martina Hrast
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Gregor Bajc
- Biotechnical Faculty, Department of Biology, University of Ljubljana, Ljubljana, Slovenia
| | - Rok Frlan
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Anže Meden
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Ljubljana, Ljubljana, Slovenia
| | - Matej Butala
- Biotechnical Faculty, Department of Biology, University of Ljubljana, Ljubljana, Slovenia
| | - Stanislav Gobec
- Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Ljubljana, Ljubljana, Slovenia,CONTACT Stanislav Gobec Faculty of Pharmacy, Department of Pharmaceutical Chemistry, University of Ljubljana, Askerceva 7, 1000Ljubljana, Slovenia
| |
Collapse
|
50
|
Balakina A, Gadomsky S, Kokovina T, Sashenkova T, Mishchenko D, Terentiev A. New Derivatives of N-Hydroxybutanamide: Preparation, MMP Inhibition, Cytotoxicity, and Antitumor Activity. Int J Mol Sci 2023; 24:16360. [PMID: 38003553 PMCID: PMC10671431 DOI: 10.3390/ijms242216360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/12/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Using a novel method of N-substituted succinimide ring opening, new N-hydroxybutanamide derivatives were synthesized. These compounds were evaluated for their ability to inhibit matrix metalloproteinases (MMPs) and their cytotoxicity. The iodoaniline derivative of N1-hydroxy-N4-phenylbutanediamide showed the inhibition of MMP-2, MMP-9, and MMP-14 with an IC50 of 1-1.5 μM. All the compounds exhibited low toxicity towards carcinoma cell lines HeLa and HepG2. The iodoaniline derivative was also slightly toxic to glioma cell lines A-172 and U-251 MG. Non-cancerous FetMSC and Vero cells were found to be the least sensitive to all the compounds. In vivo studies demonstrated that the iodoaniline derivative of N1-hydroxy-N4-phenylbutanediamide had low acute toxicity. In a mouse model of B16 melanoma, this compound showed both antitumor and antimetastatic effects, with a 61.5% inhibition of tumor growth and an 88.6% inhibition of metastasis. Our findings suggest that the iodoaniline derivative of N1-hydroxy-N4-phenylbutanediamide has potential as a lead structure for the development of new MMP inhibitors. Our new synthetic approach can be a cost-effective method for the synthesis of inhibitors of metalloenzymes with promising antitumor potential.
Collapse
Affiliation(s)
- Anastasia Balakina
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, 142432 Chernogolovka, Russia; (A.B.); (S.G.); (T.K.); (T.S.); (D.M.)
| | - Svyatoslav Gadomsky
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, 142432 Chernogolovka, Russia; (A.B.); (S.G.); (T.K.); (T.S.); (D.M.)
| | - Tatyana Kokovina
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, 142432 Chernogolovka, Russia; (A.B.); (S.G.); (T.K.); (T.S.); (D.M.)
- Faculty of Fundamental Physical-Chemical Engineering of M.V. Lomonosov MSU, Leninskie Gory, 119991 Moscow, Russia
| | - Tatyana Sashenkova
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, 142432 Chernogolovka, Russia; (A.B.); (S.G.); (T.K.); (T.S.); (D.M.)
| | - Denis Mishchenko
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, 142432 Chernogolovka, Russia; (A.B.); (S.G.); (T.K.); (T.S.); (D.M.)
- Faculty of Fundamental Physical-Chemical Engineering of M.V. Lomonosov MSU, Leninskie Gory, 119991 Moscow, Russia
- Scientific and Educational Center in Chernogolovka, State University of Education, 141014 Mytishchi, Russia
| | - Alexei Terentiev
- Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry RAS, 142432 Chernogolovka, Russia; (A.B.); (S.G.); (T.K.); (T.S.); (D.M.)
- Faculty of Fundamental Physical-Chemical Engineering of M.V. Lomonosov MSU, Leninskie Gory, 119991 Moscow, Russia
- Scientific and Educational Center in Chernogolovka, State University of Education, 141014 Mytishchi, Russia
| |
Collapse
|