1
|
Contreras M, Rafael M, Sobrino I, Almazán C, Pastor Comín JJ, Valdés JJ, Prudencio CR, de Lima Neto DF, Borin VA, Agarwal PK, Kasaija PD, Fernández-Melgar R, Rutaisire J, de la Fuente J. Modelling protein-protein interactions for the design of vaccine chimeric antigens with protective epitopes. PLoS One 2025; 20:e0318439. [PMID: 39928697 PMCID: PMC11809815 DOI: 10.1371/journal.pone.0318439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 01/15/2025] [Indexed: 02/12/2025] Open
Abstract
Ticks and tick-borne diseases are a growing burden worldwide and vaccines are effective control interventions. Vaccine formulations with tick antigens such as BM86/BM95 (BM) and Subolesin (SUB) have shown reduction in tick fitness and infestation in immunized hosts. However, antigen combination is a challenging approach to improve vaccine efficacy (E) against multiple tick species. Herein, in silico and in music algorithms were integrated to model BM-SUB protein-protein interactions to apply a quantum vaccinology approach for combining protective epitopes or immunological quantum in the chimeric antigen Q38-95. Cattle immunized with Q38-95 and infested with African blue tick Rhipicephalus decoloratus showed an 82% E similar to BM86 and higher than SUB. The immune mechanisms activated in cattle in response to vaccination with Q38-95 were mediated by anti-BM/SUB antibodies that interfered with BM-SUB interactions and through activation of other innate and adaptive immune pathways. The results support modelling protein-protein interactions affecting E to identify and combine candidate protective epitopes in chimeric antigens.
Collapse
Affiliation(s)
- Marinela Contreras
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC), Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Castilla-La Mancha (UCLM)-Junta de Comunidades de Castilla-La Mancha (JCCM), Ciudad Real, Spain
| | - Marta Rafael
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC), Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Castilla-La Mancha (UCLM)-Junta de Comunidades de Castilla-La Mancha (JCCM), Ciudad Real, Spain
| | - Isidro Sobrino
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC), Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Castilla-La Mancha (UCLM)-Junta de Comunidades de Castilla-La Mancha (JCCM), Ciudad Real, Spain
| | - Consuelo Almazán
- Laboratorio de Inmunología y Vacunas, Facultad de Ciencias Naturales, Universidad Autónoma de Querétaro, Juriquilla, Querétaro, Mexico
| | - Juan J. Pastor Comín
- Centro de Investigación y Documentación Musical CIDoM-UCLM-CSIC, Facultad de Educación de Ciudad Real, Ciudad Real, Spain
| | - James J. Valdés
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czech Republic
| | - Carlos Roberto Prudencio
- Immunology Center, Adolfo Lutz Institute, São Paulo, SP, Brazil
- Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, Brazil
| | - Daniel Ferreira de Lima Neto
- General Coordination of Public Health Laboratories, Health Surveillance Secretariat, Ministry of Health, Brasília, Brazil
| | - Veniamin A. Borin
- Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Pratul K. Agarwal
- Department of Physiological Sciences, Oklahoma State University, Stillwater, Oklahoma, United States of America
- High-Performance Computing Center, Oklahoma State University, Stillwater, Oklahoma, United States of America
| | - Paul D. Kasaija
- National Livestock Resources Research Institute (NaLIRRI/NARO), Kampala, Uganda
| | - Rubén Fernández-Melgar
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC), Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Castilla-La Mancha (UCLM)-Junta de Comunidades de Castilla-La Mancha (JCCM), Ciudad Real, Spain
| | - Justus Rutaisire
- National Livestock Resources Research Institute (NaLIRRI/NARO), Kampala, Uganda
| | - José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos (IREC), Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Castilla-La Mancha (UCLM)-Junta de Comunidades de Castilla-La Mancha (JCCM), Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, Oklahoma, United States of America
| |
Collapse
|
2
|
Su Y, Shi Y, Lai W. Revealing the Monooxygenase Mechanism for Selective Ring Cleavage of Anthraquinone by BTG13 through Multiscale Simulations. Chembiochem 2025; 26:e202400953. [PMID: 39807705 DOI: 10.1002/cbic.202400953] [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: 11/22/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/16/2025]
Abstract
BTG13, a non-heme iron-dependent enzyme with a distinctive coordination environment of four histidines and a carboxylated lysine, has been found to catalyze the cleavage of the C4a-C10 bond in anthraquinone. Contrary to typical dioxygenase mechanisms, our quantum mechanical/molecular mechanical (QM/MM) calculations reveal that BTG13 functions more like a monooxygenase. It selectively inserts an oxygen atom into the C10-C4a bond, creating a lactone species that subsequently undergoes hydrolysis, leading to the formation of a ring-opened product. This discovery highlights the unique catalytic properties of BTG13 and expands our understanding of non-heme iron enzyme mechanisms.
Collapse
Affiliation(s)
- Yanzhuang Su
- Key Laboratory of Advanced Light Conversion Materials and Biophotonics, School of Chemistry and Life Resources, Renmin University of China, Beijing, 100872, China
| | - Yusheng Shi
- Key Laboratory of Advanced Light Conversion Materials and Biophotonics, School of Chemistry and Life Resources, Renmin University of China, Beijing, 100872, China
| | - Wenzhen Lai
- Key Laboratory of Advanced Light Conversion Materials and Biophotonics, School of Chemistry and Life Resources, Renmin University of China, Beijing, 100872, China
| |
Collapse
|
3
|
Chen C, Zhang Z, Duan M, Wu Q, Yang M, Jiang L, Liu M, Li C. Aromatic-aromatic interactions drive fold switch of GA95 and GB95 with three residue difference. Chem Sci 2025; 16:1885-1893. [PMID: 39720130 PMCID: PMC11665817 DOI: 10.1039/d4sc04951a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 12/17/2024] [Indexed: 12/26/2024] Open
Abstract
Proteins typically adopt a single fold to carry out their function, but metamorphic proteins, with multiple folding states, defy this norm. Deciphering the mechanism of conformational interconversion of metamorphic proteins is challenging. Herein, we employed nuclear magnetic resonance (NMR), circular dichroism (CD), and all-atom molecular dynamics (MD) simulations to elucidate the mechanism of fold switching in proteins GA95 and GB95, which share 95% sequence homology. The results reveal that long-range interactions, especially aromatic π-π interactions involving residues F52, Y45, F30, and Y29, are critical for the protein switching from a 3α to a 4β + α fold. This study contributes to understanding how proteins with highly similar sequences fold into distinct conformations and may provide valuable insights into the protein folding code.
Collapse
Affiliation(s)
- Chen Chen
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement, Chinese Academy of Sciences Wuhan 430071 China
- Graduate University of Chinese Academy of Sciences Beijing 100049 China
| | - Zeting Zhang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement, Chinese Academy of Sciences Wuhan 430071 China
- Graduate University of Chinese Academy of Sciences Beijing 100049 China
| | - Mojie Duan
- Interdisciplinary Institute of NMR and Molecular Sciences, School of Chemistry and Chemical Engineering, The State Key Laboratory of Refractories and Metallurgy, Wuhan University of Science and Technology Wuhan 430081 China
| | - Qiong Wu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement, Chinese Academy of Sciences Wuhan 430071 China
| | - Minghui Yang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement, Chinese Academy of Sciences Wuhan 430071 China
- Graduate University of Chinese Academy of Sciences Beijing 100049 China
| | - Ling Jiang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement, Chinese Academy of Sciences Wuhan 430071 China
- Graduate University of Chinese Academy of Sciences Beijing 100049 China
| | - Maili Liu
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement, Chinese Academy of Sciences Wuhan 430071 China
- Graduate University of Chinese Academy of Sciences Beijing 100049 China
| | - Conggang Li
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan National Laboratory for Optoelectronics, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement, Chinese Academy of Sciences Wuhan 430071 China
- Graduate University of Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
4
|
Hasse T, Zhang Z, Huang YMM. In silico discovery of potential inhibitors targeting the MEIG1-PACRG complex for male contraceptive development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.16.628759. [PMID: 39763986 PMCID: PMC11702573 DOI: 10.1101/2024.12.16.628759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The interaction between meiosis-expressed gene 1 (MEIG1) and Parkin co-regulated gene (PACRG) is a critical determinant of spermiogenesis, the process by which round spermatids mature into functional spermatozoa. Disruption of the MEIG1-PACRG complex can impair sperm development, highlighting its potential as a therapeutic target for addressing male infertility or for the development of non-hormonal contraceptive methods. This study used virtual screening, molecular docking, and molecular dynamics (MD) simulations to identify small molecule inhibitors targeting the MEIG1-PACRG interface. MD simulations provided representative protein conformations, which were used to virtually screen a library of over 800,000 compounds, resulting in 48 high-ranking candidates for each protein. PACRG emerged as a favorable target due to its flexible binding pockets and better docking scores compared to MEIG1. Key binding residues with compounds included W50, Y68, N70, and E74 on MEIG1, and K93, W96, E101, and H137 on PACRG. MD simulations revealed that compound stability in MEIG1 complexes is primarily maintained by hydrogen bonding with E74 and π-π stacking interactions with W50 and Y68. In PACRG complexes, compound stabilization is facilitated by hydrogen bonding with E101 and π-π interactions involving W96 and H137. These findings highlight distinct molecular determinants of ligand binding for each protein. Our work provides mechanistic insights and identifies promising compounds for further experimental validation, establishing a foundation for developing MEIG1-PACRG interaction inhibitors as male contraceptives.
Collapse
|
5
|
Abraham CB, Lewkowicz E, Gursky O, Straub JE. Elucidating the Mechanism of Recognition and Binding of Heparin to Amyloid Fibrils of Serum Amyloid A. Biochemistry 2024. [PMID: 39688935 DOI: 10.1021/acs.biochem.4c00529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Amyloid diseases feature pathologic deposition of normally soluble proteins and peptides as insoluble fibrils in vital organs. Amyloid fibrils co-deposit with various nonfibrillar components including heparan sulfate (HS), a glycosaminoglycan that promotes amyloid formation in vitro for many unrelated proteins. HS-amyloid interactions have been proposed as a therapeutic target for inflammation-linked amyloidosis wherein N-terminal fragments of serum amyloid A (SAA) protein deposit in the kidney and liver. The structural basis for these interactions is unclear. Here, we exploit the high-resolution cryoelectron microscopy (cryo-EM) structures of ex vivo murine and human SAA fibrils in a computational study employing molecular docking, Brownian dynamics simulations, and molecular dynamics simulations to elucidate how heparin, a highly sulfated HS mimetic, recognizes and binds to amyloid protein fibrils. Our results demonstrate that negatively charged heparin chains bind to linear arrays of uncompensated positively charged basic residues along the spines of amyloid fibrils facilitated by electrostatic steering. The predicted heparin binding sites match the location of unidentified densities observed in cryo-EM maps of SAA amyloids, suggesting that these extra densities represent bound HS. Since HS is constitutively found in various amyloid deposits, our results suggest a common mechanism for HS-amyloid recognition and binding.
Collapse
Affiliation(s)
- Conor B Abraham
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Emily Lewkowicz
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, United States
| | - Olga Gursky
- Department of Pharmacology, Physiology & Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts 02118, United States
| | - John E Straub
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| |
Collapse
|
6
|
Sangavi P, Nagarajan H, Subramaniyan S, Jeyaraman J, Langeswaran K. Unveiling the oncological inhibition of bioactive compounds from Adansonia digitata via in silico analysis by targeting γ-butyrobetaine dioxygenase 1 against triple negative breast cancer. J Biomol Struct Dyn 2024:1-24. [PMID: 39660540 DOI: 10.1080/07391102.2024.2437528] [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: 03/02/2024] [Accepted: 05/15/2024] [Indexed: 12/12/2024]
Abstract
Adansonia digitata extracts are well known for their wide range of nutritional and medicinal benefits, including anti-diabetic, anti-inflammatory, antioxidant, and anti-cancerous properties. Yet, its efficacy against breast cancer has not been well-studied so far. Hence this study aims to investigate the anti-cancer properties of phytochemicals from the bark extract of the Adansonia digitata tree against BBOX1, a protein that stimulates the growth of Triple Negative Breast Cancer (TNBC) cells. TNBC is a highly aggressive and fatal form of cancer with limited therapeutic options available. By incorporating computational bioinformatics including Molecular docking, MMGBSA/PBSA, Molecular dynamics, and PCA/FEL analysis, the phytocompounds were scrutinized against BBOX1. Among 274 Phytocompounds only 37 compounds with good pharmacokinetic profiles based on ADME analysis were selected and docked with BBOX1. Of these compounds, the top 6 phytocompounds (CID_22217550, CID_559476, CID_6423866, CID_595387, CID_550931, and CID_559495) demonstrated good binding affinity, with better docking scores ranging from -8.599 to -7.207 kcal/mol respectively. Furthermore, based on MM/GBSA, Interaction profiling, and DFT analysis, only three phytocompounds namely CID_22217550, CID_559476, and CID_550931 were found to interact with the key residues such as Tyr_177, Trp_181, Asp_191, and Tyr_366 with better binding efficacy. In addition, these compounds were also observed to have the least RMS deviations with stable H-bond interactions maintained throughout the MD production run. Henceforth, the overall analysis infers that the phytocompounds CID_22217550, CID_559476, and CID_550931 shall act as potent inhibitors of BBOX1. However, their inhibitory efficacy has be to analyzed with further in vitro and in vivo analysis.
Collapse
Affiliation(s)
- P Sangavi
- Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Hemavathy Nagarajan
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Sneha Subramaniyan
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Jeyakanthan Jeyaraman
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - K Langeswaran
- Department of Biomedical Science, Alagappa University, Karaikudi, Tamil Nadu, India
| |
Collapse
|
7
|
Song X, Zhao Q, Dang M, Hou X, Liu S, Ma Z, Ren Y. Quenching and enhancement mechanisms of a novel Cd-based coordination polymer as a multiresponsive fluorescent sensor for nitrobenzene and aniline. Anal Chim Acta 2024; 1316:342865. [PMID: 38969412 DOI: 10.1016/j.aca.2024.342865] [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: 05/16/2024] [Accepted: 06/10/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Nitroaromatic compounds are inherently hazardous and explosive, so convenient and rapid detection strategies are needed for the sake of human health and the environment. There is an urgent demand for chemical sensing materials that offer high sensitivity, operational simplicity, and recognizability to effectively monitor nitroaromatic residues in industrial wastewater. Despite its importance, the mechanisms underlying fluorescence quenching or enhancement in fluorescent sensing materials have not been extensively researched. The design and synthesis of multiresponsive fluorescent sensing materials have been a great challenge until now. RESULTS In this study, a one-dimensional Cd-based fluorescent porous coordination polymer (Cd-CIP-1) was synthesized using 5-(4-cyanobenzyl)isophthalic acid (5-H2CIP) and 4,4'-bis(1-imidazolyl)biphenyl (4,4'-bimp) and used for the selective detection of nitrobenzene in aqueous solution by fluorescence quenching, with a limit of detection of 1.38 × 10-8 mol L-1. The presence of aniline in the Cd-CIP-1 solution leads to the enhancement of fluorescence property. Density functional theory and time-dependent density functional theory calculations were carried out to elucidate the mechanisms of the fluorescence changes. This study revealed that the specific pore size of Cd-CIP-1 facilitates analyte screening and enhances host-guest electron coupling. Furthermore, π-π interactions and hydrogen bond between Cd-CIP-1 and the analytes result in intermolecular orbital overlap and thereby boosting electron transfer efficiency. The different electron flow directions in NB@Cd-CIP-1 and ANI@Cd-CIP-1 lead to fluorescence quenching and enhancement. SIGNIFICANCE AND NOVELTY The multiresponsive coordination polymer (Cd-CIP-1) can selectively detect nitrobenzene and recognize aniline in aqueous solutions. The mechanism of fluorescence quenching and enhancement has been thoroughly elucidated through a combination of density functional theory and experimental approaches. This study presents a promising strategy for the practical implementation of a multiresponsive fluorescent chemical sensor.
Collapse
Affiliation(s)
- Xiaoming Song
- Shaanxi Key Laboratory of Chemical Reaction Engineering, Laboratory of New Energy and New Function Materials, College of Chemistry and Chemical Engineering, Yan'an University, Yan'an, Shaanxi, 716000, China
| | - Qingxia Zhao
- Shaanxi Key Laboratory of Chemical Reaction Engineering, Laboratory of New Energy and New Function Materials, College of Chemistry and Chemical Engineering, Yan'an University, Yan'an, Shaanxi, 716000, China
| | - Mingxuan Dang
- Shaanxi Key Laboratory of Chemical Reaction Engineering, Laboratory of New Energy and New Function Materials, College of Chemistry and Chemical Engineering, Yan'an University, Yan'an, Shaanxi, 716000, China
| | - Xiufang Hou
- Shaanxi Key Laboratory of Chemical Reaction Engineering, Laboratory of New Energy and New Function Materials, College of Chemistry and Chemical Engineering, Yan'an University, Yan'an, Shaanxi, 716000, China.
| | - Shuai Liu
- Shaanxi Key Laboratory of Chemical Reaction Engineering, Laboratory of New Energy and New Function Materials, College of Chemistry and Chemical Engineering, Yan'an University, Yan'an, Shaanxi, 716000, China
| | - Zhihu Ma
- Shaanxi Key Laboratory of Chemical Reaction Engineering, Laboratory of New Energy and New Function Materials, College of Chemistry and Chemical Engineering, Yan'an University, Yan'an, Shaanxi, 716000, China
| | - Yixia Ren
- Shaanxi Key Laboratory of Chemical Reaction Engineering, Laboratory of New Energy and New Function Materials, College of Chemistry and Chemical Engineering, Yan'an University, Yan'an, Shaanxi, 716000, China.
| |
Collapse
|
8
|
Thawani R, Repetto M, Keddy C, Nicholson K, Jones K, Nusser K, Beach CZ, Harada G, Drilon A, Davare MA. TKI type switching overcomes ROS1 L2086F in ROS1 fusion-positive cancers. NPJ Precis Oncol 2024; 8:175. [PMID: 39117775 PMCID: PMC11310217 DOI: 10.1038/s41698-024-00663-1] [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: 01/12/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024] Open
Abstract
The grammar in this abstract is generally correct, but there's a minor issue with sentence structure in one part. Here's a slightly revised version with improved grammar and flow:ROS1 tyrosine kinase inhibitors (TKIs) are highly effective in ROS1-positive non-small cell lung cancer, but resistance remains a challenge. We investigated the activity of various TKIs against wildtype and mutant ROS1, focusing on the emerging L2086F resistance mutation. Using Ba/F3 and NIH3T3 cell models, CRISPR/Cas9-edited isogenic wildtype and mutant patient-derived cell lines, and in vivo tumor growth studies, we compared type I TKIs (crizotinib, entrectinib, taletrectinib, lorlatinib, and repotrectinib) to type II TKIs (cabozantinib and merestinib) and the type I FLT3 inhibitor gilteritinib. The ROS1 L2086F mutant kinase showed resistance to type I TKIs, while type II TKIs retained activity. Gilteritinib inhibited both wildtype and L2086F mutant ROS1 but was ineffective against the G2032R mutation. Structural analyses revealed distinct binding poses for cabozantinib and gilteritinib, explaining their efficacy against L2086F. Clinical cases demonstrated cabozantinib's effectiveness in patients with TKI-resistant, ROS1 L2086F mutant NSCLCs. This study provides the first comprehensive report of ROS1 L2086F in the context of later-generation TKIs, including macrocyclic inhibitors. While cabozantinib effectively inhibits ROS1 L2086F, its multi-kinase inhibitor nature highlights the need for more selective and better-tolerated TKIs to overcome kinase-intrinsic resistance. Gilteritinib may offer an alternative for targeting ROS1 L2086F with distinct off-target toxicities, but further studies are required to fully evaluate its potential in this setting.
Collapse
Affiliation(s)
- Rajat Thawani
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Matteo Repetto
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Clare Keddy
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Katelyn Nicholson
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Kristen Jones
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Kevin Nusser
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Catherine Z Beach
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Guilherme Harada
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Alexander Drilon
- Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, 10065, USA.
- Weill Cornell Medical College, New York, NY, USA.
| | - Monika A Davare
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, 97239, USA.
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
9
|
Del Conte A, Camagni GF, Clementel D, Minervini G, Monzon AM, Ferrari C, Piovesan D, Tosatto SE. RING 4.0: faster residue interaction networks with novel interaction types across over 35,000 different chemical structures. Nucleic Acids Res 2024; 52:W306-W312. [PMID: 38686797 PMCID: PMC11223866 DOI: 10.1093/nar/gkae337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/09/2024] [Accepted: 04/19/2024] [Indexed: 05/02/2024] Open
Abstract
Residue interaction networks (RINs) are a valuable approach for representing contacts in protein structures. RINs have been widely used in various research areas, including the analysis of mutation effects, domain-domain communication, catalytic activity, and molecular dynamics simulations. The RING server is a powerful tool to calculate non-covalent molecular interactions based on geometrical parameters, providing high-quality and reliable results. Here, we introduce RING 4.0, which includes significant enhancements for identifying both covalent and non-covalent bonds in protein structures. It now encompasses seven different interaction types, with the addition of π-hydrogen, halogen bonds and metal ion coordination sites. The definitions of all available bond types have also been refined and RING can now process the complete PDB chemical component dictionary (over 35000 different molecules) which provides atom names and covalent connectivity information for all known ligands. Optimization of the software has improved execution time by an order of magnitude. The RING web server has been redesigned to provide a more engaging and interactive user experience, incorporating new visualization tools. Users can now visualize all types of interactions simultaneously in the structure viewer and network component. The web server, including extensive help and tutorials, is available from URL: https://ring.biocomputingup.it/.
Collapse
Affiliation(s)
- Alessio Del Conte
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Giorgia F Camagni
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Damiano Clementel
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | | | | - Carlo Ferrari
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Damiano Piovesan
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | | |
Collapse
|
10
|
Shyam M, Thakur A, Velez C, Daniel C, Acevedo O, Bhakta S, Jayaprakash V. Mycobactin analogue interacting with siderophore efflux-pump protein: insights from molecular dynamics simulations and whole-cell assays. FRONTIERS IN ANTIBIOTICS 2024; 3:1362516. [PMID: 39816270 PMCID: PMC11731696 DOI: 10.3389/frabi.2024.1362516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 04/16/2024] [Indexed: 01/18/2025]
Abstract
Introduction In response to continued public health emergency of antimicrobial resistance (AMR), a significant key strategy is the discovery of novel mycobacterial efflux-pump inhibitors (EPIs) as potential adjuvants in combination drug therapy. Interest in identifying new chemotypes which could potentially synergize with the existing antibiotics and can be deployed as part of a combination therapy. This strategy could delay the emergence of resistance to existing antibiotics and increase their efficacy against resistant strains of mycobacterial species. In recent decades, notable approaches have been accounted for EPI development and have resulted in the discovery of several EPIs including SQ109 and AU1235. In context, to accelerate newer EPIs with novel mode of action here we have discussed mycobactin analogues and highlighted in silico binding orientation with siderophore efflux-pump proteins MmpL4/5. Methods 3-(2-hydroxyphenyl)-5-(aryl)-pyrazoline series was investigated for whole-cell efflux-pump inhibitory activity against Mycobacterium smegmatis and Mycobacterium abscessus. Machine learning and molecular dynamics were performed to construct a MmpL4/5 complex embedded in a lipid bilayer to identify the putative binding site and to predict ligand-protein binding energetics. Furthermore, the identified HIT compound was investigated in synergistic assay with bedaquiline. Results Compound Il, 2-(5-(4-fluorophenyl)-4,5-dihydro-1H-pyrazol-3-yl)phenol, was identified as the most potent efflux pump inhibitor against M. smegmatis in whole-cell efflux-pump investigation. Followed HIT Il employed against M. abscessus for efflux-pump inhibition investigations and notable whole-cell efflux-pump inhibitory profile has been observed. The theoretical investigations predicted compound Il to be selective towards MmpL4, with significant hydrogen bonding and π-π stacking interactions effectively blocking a critical Asp-Tyr dyad interaction network necessary for proton translocation. Compound Il with bedaquiline highlighted an additive profile against the M. abscessus pathogen. Conclusions MD simulations and whole-cell assays are indicating potential development of compound Il as an adjunct to the existing therapeutic regimen against mycobacterial infections.
Collapse
Affiliation(s)
- Mousumi Shyam
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Ranchi, India
- Mycobacteria Research Laboratory, Department of Natural Sciences, Institute of Structural and Molecular Biology, Birkbeck, University of London, London, United Kingdom
| | - Abhishek Thakur
- Department of Chemistry, University of Miami, Coral Gables, FL, United States
| | - Caroline Velez
- Department of Chemistry, University of Miami, Coral Gables, FL, United States
| | - Chris Daniel
- Mycobacteria Research Laboratory, Department of Natural Sciences, Institute of Structural and Molecular Biology, Birkbeck, University of London, London, United Kingdom
| | - Orlando Acevedo
- Department of Chemistry, University of Miami, Coral Gables, FL, United States
| | - Sanjib Bhakta
- Mycobacteria Research Laboratory, Department of Natural Sciences, Institute of Structural and Molecular Biology, Birkbeck, University of London, London, United Kingdom
| | - Venkatesan Jayaprakash
- Department of Pharmaceutical Sciences & Technology, Birla Institute of Technology, Ranchi, India
| |
Collapse
|
11
|
Wang Z, Li Y, Sun M. The effect of weak π-π interactions on single-molecule electron transport properties of the tetraphenylethene molecule and its derivatives: a first-principles study. Phys Chem Chem Phys 2024; 26:1067-1076. [PMID: 38095244 DOI: 10.1039/d3cp04593e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
Intramolecular π-π interactions are a significant research focus in fields such as chemistry, biology, and materials science. Different configurations of benzene-benzene moieties within a molecule can affect the magnitude of their π-π interactions, consequently influencing the electronic transport capabilities of the molecule. In this study, we designed three π-conjugated molecules, TPEM, TPEEM, and TEEPM, based on tetraphenylethene (TPE). These three molecules exhibit three distinct π-conjugated structures: linear cis-π-conjugation, linear trans-π-conjugation, and cross-π-conjugation. Thereinto, TPEM and TPEEM molecules share the same TPE core, with identical π-π interaction distances, while the TEEPM molecule has acetylene groups between the TPE units, thereby increasing the π-π interaction distances between the benzene moieties. Using density functional theory calculations combined with non-equilibrium Green's function (DFT+NEGF), our results reveal that the conductance order of different π-conjugated structures in TPEM and TPEEM molecules is as follows: cis > cross ≈ trans. Through analysis of transmission spectra, transmission pathways, and the innermost π orbitals, we find that in TPEM and TPEEM molecules, the cis- and cross-π-conjugated structures exhibit π-π interactions between benzene moieties and provide special through-space electron transport pathways, enhancing their electronic transport capabilities in coordination with the bonded molecular framework, whereas their trans-conjugated structures only allow electron transport along the molecular backbone. In contrast, in TEEPM molecule, due to the absence of π-π interactions, the conductance of different π-conjugated structures is primarily determined by the molecular backbone and follows the order: trans > cis > cross. These findings provide a theoretical basis for designing single-molecule electronic devices with multiple electron channels based on intramolecular π-π interactions.
Collapse
Affiliation(s)
- Zhiye Wang
- Department of Materials and Metallurgy, Wuhan University of Science and Technology, 430081, Wuhan, China.
| | - Yunchuan Li
- Department of Materials and Metallurgy, Wuhan University of Science and Technology, 430081, Wuhan, China.
| | - Mingjun Sun
- Department of Materials and Metallurgy, Wuhan University of Science and Technology, 430081, Wuhan, China.
| |
Collapse
|
12
|
Hernández-Rocamora VM, Molina R, Alba A, Carrasco-López C, Rojas-Altuve A, Panjikar S, Medina A, Usón I, Alfonso C, Galán B, Rivas G, Hermoso JA, Sanz JM. Structural characterization of PaaX, the main repressor of the phenylacetate degradation pathway in Escherichia coli W: A novel fold of transcription regulator proteins. Int J Biol Macromol 2024; 254:127935. [PMID: 37949283 DOI: 10.1016/j.ijbiomac.2023.127935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/30/2023] [Accepted: 11/05/2023] [Indexed: 11/12/2023]
Abstract
PaaX is a transcriptional repressor of the phenylacetic acid (PAA) catabolic pathway, a central route for bacterial aerobic degradation of aromatic compounds. Induction of the route is achieved through the release of PaaX from its promoter sequences by the first compound of the pathway, phenylacetyl-coenzyme A (PA-CoA). We report the crystal structure of PaaX from Escherichia coli W. PaaX displays a novel type of fold for transcription regulators, showing a dimeric conformation where the monomers present a three-domain structure: an N-terminal winged helix-turn-helix domain, a dimerization domain similar to the Cas2 protein and a C-terminal domain without structural homologs. The domains are separated by a crevice amenable to harbour a PA-CoA molecule. The biophysical characterization of the protein in solution confirmed several hints predicted from the structure, i.e. its dimeric conformation, a modest importance of cysteines and a high dependence of solubility and thermostability on ionic strength. At a moderately acidic pH, the protein formed a stable folding intermediate with remaining α-helical structure, a disrupted tertiary structure and exposed hydrophobic patches. Our results provide valuable information to understand the stability and mechanism of PaaX and pave the way for further analysis of other regulators with similar structural configurations.
Collapse
Affiliation(s)
- Víctor M Hernández-Rocamora
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche, Universidad Miguel Hernández, Av. Universidad, s/n, E-03202 Elche, Alicante, Spain; Centre for Bacterial Cell Biology, Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rafael Molina
- Department of Crystallography and Structural Biology, Instituto de Química-Física "Blas Cabrera", Consejo Superior de Investigaciones Científicas, Serrano 119, 28006 Madrid, Spain
| | - Alejandra Alba
- Department of Crystallography and Structural Biology, Instituto de Química-Física "Blas Cabrera", Consejo Superior de Investigaciones Científicas, Serrano 119, 28006 Madrid, Spain
| | - César Carrasco-López
- Department of Crystallography and Structural Biology, Instituto de Química-Física "Blas Cabrera", Consejo Superior de Investigaciones Científicas, Serrano 119, 28006 Madrid, Spain
| | - Alzoray Rojas-Altuve
- Department of Crystallography and Structural Biology, Instituto de Química-Física "Blas Cabrera", Consejo Superior de Investigaciones Científicas, Serrano 119, 28006 Madrid, Spain
| | - Santosh Panjikar
- Australian Synchrotron, ANSTO, Clayton, Australia; Department of Molecular Biology and Biochemistry, Monash University, Melbourne, Australia
| | - Ana Medina
- Crystallographic Methods, Institute of Molecular Biology of Barcelona (IBMB-CSIC), Baldiri Reixach 15, 08028 Barcelona, Spain
| | - Isabel Usón
- Crystallographic Methods, Institute of Molecular Biology of Barcelona (IBMB-CSIC), Baldiri Reixach 15, 08028 Barcelona, Spain; ICREA: Institució Catalana de Recerca i Estudis Avançats, Pg. Lluis Companys 23, 08010 Barcelona, Spain
| | - Carlos Alfonso
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28049 Madrid, Spain
| | - Beatriz Galán
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28049 Madrid, Spain
| | - Germán Rivas
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28049 Madrid, Spain
| | - Juan A Hermoso
- Department of Crystallography and Structural Biology, Instituto de Química-Física "Blas Cabrera", Consejo Superior de Investigaciones Científicas, Serrano 119, 28006 Madrid, Spain.
| | - Jesús M Sanz
- Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28049 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
13
|
Demircan Ozelcaglayan E, Honek JF, Parker WJ. Molecular level investigation of interactions between pharmaceuticals and β-cyclodextrin (β-CD) functionalized adsorption sites for removal of pharmaceutical contaminants from water. CHEMOSPHERE 2024; 347:140639. [PMID: 37939929 DOI: 10.1016/j.chemosphere.2023.140639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/23/2023] [Accepted: 11/05/2023] [Indexed: 11/10/2023]
Abstract
This study describes a novel application of the use of molecular modeling tools for investigating the adsorption of organic micropollutants (OMPs) from water by nanocomposites. The partitioning of pharmaceuticals onto β-Cyclodextrin (β-CD) functionalized adsorbents was investigated at the molecular level to explore the atomistic interactions of pharmaceutical contaminants in water systems with β-CD and to provide insight into possible approaches for removal of pharmaceuticals from water. Molecular electrostatic surface potential mapping of β-CD derivatives was employed to examine the impact of substitution degree of β-CD and type of grafting agent on host-guest complexation. The stability of the complexes of selected pharmaceuticals and β-CD derivatives were assessed via molecular dynamics simulations to evaluate competitive adsorption between organic micropollutants (OMPs) and between OMPs and fulvic acid, a representative natural organic material (NOM) component found in water systems. Molecular electrostatic surface potential maps showed that grafting agents with aromatic and amine functional groups were found to provide attractive interactions for negatively charged OMPs. In addition, optimization of substitution degree of β-CD is necessary to enhance adsorption of target OMPs. Furthermore, it was found that aromatic ring bearing grafting agents can provide additional electrostatic attractions by π-π interactions with the aromatic ring of the OMPs. The impact of common water quality characteristics on adsorption was assessed and it was revealed that the effect of pH and calcium on adsorption depends on the ionizable functional groups present on the grafting agent. Molecular dynamics simulations showed that adsorption of target OMPs does not solely depend on hydrophobicity but is affected by electrostatic interactions. The simulations revealed that fulvic acid which is commonly present in environmental waters can be a competitor with ibuprofen for the β-CD cavity. Ultimately, this study showed that molecular level simulation can be effectively employed to investigate adsorption of OMPs by β-CD functionalized adsorbents and could be employed to enhance their design and subsequent environmental applications.
Collapse
Affiliation(s)
- Ezgi Demircan Ozelcaglayan
- Department of Civil and Environmental Engineering, University of Waterloo, 200 University Avenue West, Waterloo, N2L 3G1, Ontario, Canada.
| | - John F Honek
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, N2L 3G1, Ontario, Canada
| | - Wayne J Parker
- Department of Civil and Environmental Engineering, University of Waterloo, 200 University Avenue West, Waterloo, N2L 3G1, Ontario, Canada
| |
Collapse
|
14
|
Chen X, Zhang T, Liu H, Zang J, Lv C, Du M, Zhao G. Shape-Anisotropic Assembly of Protein Nanocages with Identical Building Blocks by Designed Intermolecular π-π Interactions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2305398. [PMID: 37870198 PMCID: PMC10724428 DOI: 10.1002/advs.202305398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/12/2023] [Indexed: 10/24/2023]
Abstract
Protein lattices that shift the structure and shape anisotropy in response to environmental cues are closely coupled to potential functionality. However, to design and construct shape-anisotropic protein arrays from the same building blocks in response to different external stimuli remains challenging. Here, by a combination of the multiple, symmetric interaction sites on the outer surface of protein nanocages and the tunable features of phenylalanine-phenylalanine interactions, a protein engineering approach is reported to construct a variety of superstructures with shape anisotropy, including 3D cubic, 2D hexagonal layered, and 1D rod-like crystalline protein nanocage arrays by using one single protein building block. Notably, the assembly of these crystalline protein arrays is reversible, which can be tuned by external stimuli (pH and ionic strength). The anisotropic morphologies of the fabricated macroscopic crystals can be correlated with the Å-to-nm scale protein arrangement details by crystallographic elucidation. These results enhance the understanding of the freedom offered by an object's symmetry and inter-object π-π stacking interactions for protein building blocks to assemble into direction- and shape-anisotropic biomaterials.
Collapse
Affiliation(s)
- Xuemin Chen
- College of Food Science & Nutritional EngineeringBeijing Key Laboratory of Functional Food from Plant ResourcesChina Agricultural UniversityBeijing100083China
| | - Tuo Zhang
- College of Food Science & Nutritional EngineeringBeijing Key Laboratory of Functional Food from Plant ResourcesChina Agricultural UniversityBeijing100083China
| | - Hanxiong Liu
- School of Food Science and TechnologyNational Engineering Research Center of SeafoodDalian Polytechnic UniversityDalian116034China
| | - Jiachen Zang
- College of Food Science & Nutritional EngineeringBeijing Key Laboratory of Functional Food from Plant ResourcesChina Agricultural UniversityBeijing100083China
| | - Chenyan Lv
- College of Food Science & Nutritional EngineeringBeijing Key Laboratory of Functional Food from Plant ResourcesChina Agricultural UniversityBeijing100083China
| | - Ming Du
- School of Food Science and TechnologyNational Engineering Research Center of SeafoodDalian Polytechnic UniversityDalian116034China
| | - Guanghua Zhao
- College of Food Science & Nutritional EngineeringBeijing Key Laboratory of Functional Food from Plant ResourcesChina Agricultural UniversityBeijing100083China
| |
Collapse
|
15
|
Shen Y, Xiao Y, Edkins RM, Youngs TGA, Hughes TL, Tellam J, Edkins K. Elucidating the hydrotropism behaviour of aqueous caffeine and sodium benzoate solution through NMR and neutron total scattering analysis. Int J Pharm 2023; 647:123520. [PMID: 37858637 DOI: 10.1016/j.ijpharm.2023.123520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/21/2023]
Abstract
Hydrotropism is a convenient way to increase the solubility of drugs by up to several orders of magnitude, and even though it has been researched for decades with both experimental and simulation methods, its mechanism is still unknown. Here, we use caffeine/sodium benzoate (CAF-SB) as model system to explore the behaviour of caffeine solubility enhancement in water through NMR spectroscopy and neutron total scattering. 1H NMR shows strong interaction between caffeine and sodium benzoate in water. Neutron total scattering combined with empirical potential structure refinement, a systematic method to study the solution structure, reveals π-stacking between caffeine and the benzoate anion as well as Coulombic interactions with the sodium cation. The strongest hydrogen bond interaction in the system is between benzoate and water, which help dissolve CAF-SB complex and increase the solubility of CAF in water. Besides, the stronger interaction between CAF and water and the distortion of water structure are further mechanisms of the CAF solubility enhancement. It is likely that the variety of mechanisms for hydrotropism shown in this system can be found for other hydrotropes, and NMR spectroscopy and neutron total scattering can be used as complementary techniques to generate a holistic picture of hydrotropic solutions.
Collapse
Affiliation(s)
- Yichun Shen
- School of Health Sciences, University of Manchester, Stopford Building, Oxford Road, Manchester M13 9PT, UK
| | - Yitian Xiao
- School of Health Sciences, University of Manchester, Stopford Building, Oxford Road, Manchester M13 9PT, UK
| | - Robert M Edkins
- Department of Pure and Applied Chemistry, University of Strathclyde, Thomas Graham Building, 295, Cathedral Street, Glasgow, G1 1XL, UK
| | - Tristan G A Youngs
- ISIS Pulsed Neutron and Muon Source, STFC Rutherford Appleton Laboratory, Harwell Oxford, Didcot, OX11 0QX, UK
| | - Terri-Louise Hughes
- ISIS Pulsed Neutron and Muon Source, STFC Rutherford Appleton Laboratory, Harwell Oxford, Didcot, OX11 0QX, UK
| | - James Tellam
- ISIS Deuteration Facility, STFC Rutherford Appleton Laboratory, Harwell Oxford, Didcot, OX11 0QX, UK
| | - Katharina Edkins
- School of Health Sciences, University of Manchester, Stopford Building, Oxford Road, Manchester M13 9PT, UK.
| |
Collapse
|
16
|
Lee H, Liu X, An JP, Wang Y. Identification of Polymethoxyflavones (PMFs) from Orange Peel and Their Inhibitory Effects on the Formation of Trimethylamine (TMA) and Trimethylamine-N-oxide (TMAO) Using cntA/B and cutC/D Enzymes and Molecular Docking. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:16114-16124. [PMID: 37851928 DOI: 10.1021/acs.jafc.3c04462] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
This study investigates the inhibitory effects of polymethoxyflavones (PMFs) on enzymes involved in the production of trimethylamine (TMA) and trimethylamine-N-oxide (TMAO). PMFs were isolated from Valencia orange peel and identified using column separation and NMR techniques. The findings reveal that nobiletin and 3,6,7,8,2',5'-hexamethoxyflavone significantly suppress cntA/B and cutC/D, respectively. Furthermore, 3,6,7,8,2',5'-hexamethoxyflavone decreases the level of TMAO formation by suppressing the FMO3 mRNA level. This study elucidates that specific structural features of PMFs can contribute to their interactions with enzymes. Our study represents the first demonstration of the ability of PMFs to mitigate the risk of cardiovascular disease (CVD) by inhibiting enzymes responsible for TMA production, which are generated by gut microbiomes. Furthermore, we introduce a novel model system utilizing TMA-induced HepG2 cells to assess and compare the inhibitory effects of PMFs on TMAO production. These findings could pave the way for the development of novel therapeutic approaches to manage CVD.
Collapse
Affiliation(s)
- Hana Lee
- Department of Food Science and Human Nutrition, Citrus Research and Education Center, University of Florida, Lake Alfred, Florida 33850, United States
| | - Xin Liu
- Department of Food Science and Human Nutrition, Citrus Research and Education Center, University of Florida, Lake Alfred, Florida 33850, United States
| | - Jin-Pyo An
- Department of Food Science and Human Nutrition, Citrus Research and Education Center, University of Florida, Lake Alfred, Florida 33850, United States
| | - Yu Wang
- Department of Food Science and Human Nutrition, Citrus Research and Education Center, University of Florida, Lake Alfred, Florida 33850, United States
| |
Collapse
|
17
|
Dou T, Lian T, Shu S, He Y, Jiang J. The substrate and inhibitor binding mechanism of polyspecific transporter OAT1 revealed by high-resolution cryo-EM. Nat Struct Mol Biol 2023; 30:1794-1805. [PMID: 37845412 PMCID: PMC11406556 DOI: 10.1038/s41594-023-01123-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 09/12/2023] [Indexed: 10/18/2023]
Abstract
Organic anion transporters (OATs) of the SLC22 family have crucial roles in the transport of organic anions, including metabolites and therapeutic drugs, and in transporter-mediated drug-drug interactions. In the kidneys, OATs facilitate the elimination of metabolic waste products and xenobiotics. However, their transport activities can lead to the accumulation of certain toxic compounds within cells, causing kidney damage. Moreover, OATs are important drug targets, because their inhibition modulates the elimination or retention of substrates linked to diseases. Despite extensive research on OATs, the molecular basis of their substrate and inhibitor binding remains poorly understood. Here we report the cryo-EM structures of rat OAT1 (also known as SLC22A6) and its complexes with para-aminohippuric acid and probenecid at 2.1, 2.8 and 2.9 Å resolution, respectively. Our findings reveal a highly conserved substrate binding mechanism for SLC22 transporters, wherein four aromatic residues form a cage to accommodate the polyspecific binding of diverse compounds.
Collapse
Affiliation(s)
- Tongyi Dou
- Laboratory of Membrane Proteins and Structural Biology, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Tengfei Lian
- Laboratory of Membrane Proteins and Structural Biology, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Shi Shu
- Laboratory of Membrane Proteins and Structural Biology, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Yi He
- Fermentation Facility, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Jiansen Jiang
- Laboratory of Membrane Proteins and Structural Biology, Biochemistry and Biophysics Center, National Heart, Lung, and Blood Institute, Bethesda, MD, USA.
| |
Collapse
|
18
|
Tao Y, Xia W, Zhao Q, Xiang H, Han C, Zhang S, Gu W, Tang W, Li Y, Tan L, Li D, Liu C. Structural mechanism for specific binding of chemical compounds to amyloid fibrils. Nat Chem Biol 2023; 19:1235-1245. [PMID: 37400537 DOI: 10.1038/s41589-023-01370-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 05/26/2023] [Indexed: 07/05/2023]
Abstract
Amyloid fibril is an important pharmaceutical target for diagnostic and therapeutic treatment of neurodegenerative diseases. However, rational design of chemical compounds that interact with amyloid fibrils is unachievable due to the lack of mechanistic understanding of the ligand-fibril interaction. Here we used cryoelectron microscopy to survey the amyloid fibril-binding mechanism of a series of compounds including classic dyes, (pre)clinical imaging tracers and newly identified binders from high-throughput screening. We obtained clear densities of several compounds in complex with an α-synuclein fibril. These structures unveil the basic mechanism of the ligand-fibril interaction, which exhibits remarkable difference from the canonical ligand-protein interaction. In addition, we discovered a druggable pocket that is also conserved in the ex vivo α-synuclein fibrils from multiple system atrophy. Collectively, these findings expand our knowledge of protein-ligand interaction in the amyloid fibril state, which will enable rational design of amyloid binders in a medicinally beneficial way.
Collapse
Affiliation(s)
- Youqi Tao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China
| | - Wencheng Xia
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Qinyue Zhao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China
| | - Huaijiang Xiang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Chao Han
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Shenqing Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Gu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Wenjun Tang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Ying Li
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Li Tan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
- University of the Chinese Academy of Sciences, Beijing, China
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China.
- Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai, China.
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
- University of the Chinese Academy of Sciences, Beijing, China.
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
19
|
Yu J, Song G, Guo W, Le L, Xu F, Wang T, Wang F, Wu Y, Gu X, Pu L. ZmBELL10 interacts with other ZmBELLs and recognizes specific motifs for transcriptional activation to modulate internode patterning in maize. THE NEW PHYTOLOGIST 2023; 240:577-596. [PMID: 37583092 DOI: 10.1111/nph.19192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/15/2023] [Indexed: 08/17/2023]
Abstract
Plant height is an important agronomic trait that affects crop yield. Elucidating the molecular mechanism underlying plant height regulation is also an important question in developmental biology. Here, we report that a BELL transcription factor, ZmBELL10, positively regulates plant height in maize (Zea mays). Loss of ZmBELL10 function resulted in shorter internodes, fewer nodes, and smaller kernels, while ZmBELL10 overexpression increased plant height and hundred-kernel weight. Transcriptome analysis and chromatin immunoprecipitation followed by sequencing showed that ZmBELL10 recognizes specific sequences in the promoter of its target genes and activates cell division- and cell elongation-related gene expression, thereby influencing node number and internode length in maize. ZmBELL10 interacted with several other ZmBELL proteins via a spatial structure in its POX domain to form protein complexes involving ZmBELL10. All interacting proteins recognized the same DNA sequences, and their interaction with ZmBELL10 increased target gene expression. We identified the key residues in the POX domain of ZmBELL10 responsible for its protein-protein interactions, but these residues did not affect its transactivation activity. Collectively, our findings shed light on the functions of ZmBELL10 protein complexes and provide potential targets for improving plant architecture and yield in maize.
Collapse
Affiliation(s)
- Jia Yu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Guangshu Song
- Maize Research Institute, Jilin Academy of Agricultural Sciences, Gongzhuling, 136100, China
| | - Weijun Guo
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Liang Le
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Fan Xu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Ting Wang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
- Shangrao Normal University, Shangrao, 334001, China
| | - Fanhua Wang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Yue Wu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Xiaofeng Gu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Li Pu
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| |
Collapse
|
20
|
Dias AMGC, Moreira IP, Lychko I, Lopes Soares C, Nurrito A, Moura Barbosa AJ, Lutz-Bueno V, Mezzenga R, Carvalho AL, Pina AS, Roque ACA. Hierarchical self-assembly of a reflectin-derived peptide. Front Chem 2023; 11:1267563. [PMID: 37810582 PMCID: PMC10552760 DOI: 10.3389/fchem.2023.1267563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Reflectins are a family of intrinsically disordered proteins involved in cephalopod camouflage, making them an interesting source for bioinspired optical materials. Understanding reflectin assembly into higher-order structures by standard biophysical methods enables the rational design of new materials, but it is difficult due to their low solubility. To address this challenge, we aim to understand the molecular self-assembly mechanism of reflectin's basic unit-the protopeptide sequence YMDMSGYQ-as a means to understand reflectin's assembly phenomena. Protopeptide self-assembly was triggered by different environmental cues, yielding supramolecular hydrogels, and characterized by experimental and theoretical methods. Protopeptide films were also prepared to assess optical properties. Our results support the hypothesis for the protopeptide aggregation model at an atomistic level, led by hydrophilic and hydrophobic interactions mediated by tyrosine residues. Protopeptide-derived films were optically active, presenting diffuse reflectance in the visible region of the light spectrum. Hence, these results contribute to a better understanding of the protopeptide structural assembly, crucial for the design of peptide- and reflectin-based functional materials.
Collapse
Affiliation(s)
- Ana Margarida Gonçalves Carvalho Dias
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Inês Pimentel Moreira
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Iana Lychko
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Cátia Lopes Soares
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Arianna Nurrito
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Arménio Jorge Moura Barbosa
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Viviane Lutz-Bueno
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
- Paul Scherrer Institute, Villigen, Switzerland
| | - Raffaele Mezzenga
- Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Ana Luísa Carvalho
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Ana Sofia Pina
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Ana Cecília Afonso Roque
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
- UCIBIO—Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, Universidade NOVA de Lisboa, Caparica, Portugal
| |
Collapse
|
21
|
Li N, Shou Z, Yang S, Cheng X, Chen C, Zheng S, Shi Y, Tang H. Subtle distinction in molecular structure of flavonoids leads to vastly different coating efficiency and mechanism of metal-polyphenol networks with excellent antioxidant activities. Colloids Surf B Biointerfaces 2023; 229:113454. [PMID: 37499546 DOI: 10.1016/j.colsurfb.2023.113454] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 07/01/2023] [Accepted: 07/13/2023] [Indexed: 07/29/2023]
Abstract
Metal-polyphenol networks (MPNs) are of immense scientific interest because of their simple and rapid process to deposit on various substrates or particles with different shapes. However, there are rare reports on the effect of polyphenol molecular structure on coating efficiency and mechanism of MPNs. Herein, three typical flavonoid polyphenols, catechin (Cat), epigallocatechin (EGC) and procyanidin (PC), with the same skeleton (C6-C3-C6) but subtle distinction in molecular structure, were selected to build MPN coatings with ferric ions (Fe3+). And various techniques combined with the density functional theory (DFT) were applied to deeply reveal the roles of coordinative phenolic hydroxyl groups as well as noncovalent interactions (hydrogen bonding and π - π stacking) in the formation of flavonoid-based MPNs. We found that more accessible numbers of coordinative phenolic hydroxyl groups, the higher coating efficiency. In these flavonoid-based MPNs, the single-complex is the predominant during the coordinative modes between phenolic hydroxyl and Fe3+, not the previously reported mono-complex, bis-complex and/or tris-complex. Besides coordinative interaction, noncovalent interactions also contribute to MPNs formation, and hydrogen bonds prevail in the noncovalent interaction compared with π-π stacking. And these engineered MPN coatings can endow the substrate with excellent antioxidant activities. This study contributes to in-depth understanding the building mechanism of flavonoid-based MPNs, and increasing coating efficiency by choosing proper polyphenols.
Collapse
Affiliation(s)
- Na Li
- Department of Anesthesiology, Wenzhou Key Laboratory of perioperative medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, People's Republic of China; Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, People's Republic of China
| | - Zeyu Shou
- Department of Orthopaedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| | - Siyun Yang
- College of Science and Technology, Wenzhou-Kean University, 88 Daxue Rd, Wenzhou, Zhejiang 325060, People's Republic of China
| | - Xinxiu Cheng
- Xinjiang Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Urumqi 830011, People's Republic of China
| | - Chun Chen
- Department of Orthopaedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, People's Republic of China
| | - Shengwu Zheng
- Wenzhou Celecare Medical Instruments Co.,Ltd, Wenzhou 325000, People's Republic of China
| | - Yelu Shi
- College of Science and Technology, Wenzhou-Kean University, 88 Daxue Rd, Wenzhou, Zhejiang 325060, People's Republic of China.
| | - Hongli Tang
- Department of Anesthesiology, Wenzhou Key Laboratory of perioperative medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, People's Republic of China.
| |
Collapse
|
22
|
Fu H, Gray KA. Effect of molecular structure on the adsorption behavior of sulfanilamide antibiotics on crumpled graphene balls. WATER RESEARCH 2023; 242:120177. [PMID: 37348418 DOI: 10.1016/j.watres.2023.120177] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/06/2023] [Accepted: 06/05/2023] [Indexed: 06/24/2023]
Abstract
Since the 1930s, sulfonamide(SA)-based antibiotics have served as important pharmaceuticals, but their widespread detection in water systems threatens aquatic organisms and human health. Adsorption via graphene, its modified form (graphene oxide, GO), and related nanocomposites is a promising method to remove SAs, owing to the strong and selective surface affinity of graphene/GO with aromatic compounds. However, a deeper understanding of the mechanisms of interaction between the chemical structure of SAs and the GO surface is required to predict the performance of GO-based nanostructured materials to adsorb the individual chemicals making up this large class of pharmaceuticals. In this research, we studied the adsorptive performance of 3D crumpled graphene balls (CGBs) to remove 10 SAs and 13 structural analogs from water. The maximum adsorption capacity qm of SAs on CGB increased with the number of (1) aromatic rings; (2) electron-donating functional groups; (3) hydrogen bonding acceptor sites. Furthermore, the CGB surface displayed a preference for homocyclic relative to heterocyclic aromatic structures. A leading mechanism, π-π electron-donor-acceptor interaction, combined with hydrogen bonding, explains these trends. We developed a multiple linear regression model capable of predicting the qm as a function of SA chemical structure and properties and the oxidation level of CGB. The model predicted the adsorptive behaviors of SAs well with the exception of a chlorinated/fluorinated SA. The insights afforded by these experiments and modeling will aid in tailoring graphene-based adsorbents to remove micropollutants from water and reduce the growing public health threats associated with antibiotic resistance and endocrine-disrupting chemicals.
Collapse
Affiliation(s)
- Han Fu
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Kimberly A Gray
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
23
|
Xiang J, Tao Y, Xia Y, Luo S, Zhao Q, Li B, Zhang X, Sun Y, Xia W, Zhang M, Kang SS, Ahn EH, Liu X, Xie F, Guan Y, Yang JJ, Bu L, Wu S, Wang X, Cao X, Liu C, Zhang Z, Li D, Ye K. Development of an α-synuclein positron emission tomography tracer for imaging synucleinopathies. Cell 2023; 186:3350-3367.e19. [PMID: 37421950 PMCID: PMC10527432 DOI: 10.1016/j.cell.2023.06.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/16/2023] [Accepted: 06/07/2023] [Indexed: 07/10/2023]
Abstract
Synucleinopathies are characterized by the accumulation of α-synuclein (α-Syn) aggregates in the brain. Positron emission tomography (PET) imaging of synucleinopathies requires radiopharmaceuticals that selectively bind α-Syn deposits. We report the identification of a brain permeable and rapid washout PET tracer [18F]-F0502B, which shows high binding affinity for α-Syn, but not for Aβ or Tau fibrils, and preferential binding to α-Syn aggregates in the brain sections. Employing several cycles of counter screenings with in vitro fibrils, intraneuronal aggregates, and neurodegenerative disease brain sections from several mice models and human subjects, [18F]-F0502B images α-Syn deposits in the brains of mouse and non-human primate PD models. We further determined the atomic structure of the α-Syn fibril-F0502B complex by cryo-EM and revealed parallel diagonal stacking of F0502B on the fibril surface through an intense noncovalent bonding network via inter-ligand interactions. Therefore, [18F]-F0502B is a promising lead compound for imaging aggregated α-Syn in synucleinopathies.
Collapse
Affiliation(s)
- Jie Xiang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Neurobiology, Fourth Military Medical University, Xi'an, China
| | - Youqi Tao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China; Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yiyuan Xia
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Biomedical Sciences, School of Medicine, JiangHan University, #8, Sanjiaohu Rd., Wuhan 430056, China
| | - Shilin Luo
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Qinyue Zhao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China; Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Bowei Li
- Shenzhen Institute of Advanced Technology, University of Chinese Academy of Science, Shenzhen, Guangdong 518055, China
| | - Xiaoqian Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, China
| | - Yunpeng Sun
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Wencheng Xia
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Mingming Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Seong Su Kang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Eun-Hee Ahn
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Fang Xie
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- Department of Nuclear Medicine & PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Jenny J Yang
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | - Lihong Bu
- PET-CT/MRI Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shengxi Wu
- Department of Neurobiology, Fourth Military Medical University, Xi'an, China
| | - Xiaochuan Wang
- Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuebing Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, China
| | - Cong Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| | - Dan Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China; Zhangjiang Institute for Advanced Study, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China.
| |
Collapse
|
24
|
McLeod MJ, Tran N, McCluskey GD, Gillis TD, Bearne SL, Holyoak T. A metal-dependent conformational change provides a structural basis for the inhibition of CTP synthase by gemcitabine-5'-triphosphate. Protein Sci 2023; 32:e4648. [PMID: 37106216 PMCID: PMC10182726 DOI: 10.1002/pro.4648] [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: 03/08/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 04/29/2023]
Abstract
CTP synthases (CTPS) catalyze the de novo production of CTP using UTP, ATP, and l-glutamine with the anticancer drug metabolite gemcitabine-5'-triphosphate (dF-dCTP) being one of its most potent nucleotide inhibitors. To delineate the structural origins of this inhibition, we solved the structures of Escherichia coli CTPS (ecCTPS) in complex with CTP (2.0 Å), 2'-ribo-F-dCTP (2.0 Å), 2'-arabino-F-CTP (2.4 Å), dF-dCTP (2.3 Å), dF-dCTP and ADP (2.1 Å), and dF-dCTP and ATP (2.1 Å). These structures revealed that the increased binding affinities observed for inhibitors bearing the 2'-F-arabino group (dF-dCTP and F-araCTP), relative to CTP and F-dCTP, arise from interactions between the inhibitor's fluorine atom exploiting a conserved hydrophobic pocket formed by F227 and an interdigitating loop from an adjacent subunit (Q114-V115-I116). Intriguingly, crystal structures of ecCTPS•dF-dCTP complexes in the presence of select monovalent and divalent cations demonstrated that the in crystallo tetrameric assembly of wild-type ecCTPS was induced into a conformation similar to inhibitory ecCTPS filaments solely through the binding of Na+ -, Mg2+ -, or Mn2+ •dF-dCTP. However, in the presence of potassium, the dF-dCTP-bound structure is demetalated and in the low-affinity, non-filamentous conformation, like the conformation seen when bound to CTP and the other nucleotide analogues. Additionally, CTP can also induce the filament-competent conformation linked to high-affinity dF-dCTP binding in the presence of high concentrations of Mg2+ . This metal-dependent, compacted CTP pocket conformation therefore furnishes the binding environment responsible for the tight binding of dF-dCTP and provides insights for further inhibitor design.
Collapse
Affiliation(s)
- Matthew J. McLeod
- Department of BiologyUniversity of WaterlooWaterlooOntarioCanada
- Present address:
Laboratory of Atomic and Solid State PhysicsCornell UniversityIthacaNew YorkUSA
| | - Norman Tran
- Department of BiologyUniversity of WaterlooWaterlooOntarioCanada
| | - Gregory D. McCluskey
- Department of Biochemistry & Molecular BiologyDalhousie UniversityHalifaxNova ScotiaCanada
- Present address:
Department of Chemistry and Biomolecular SciencesUniversity of OttawaOttawaOntarioCanada
| | - Tom D. Gillis
- Department of Biochemistry & Molecular BiologyDalhousie UniversityHalifaxNova ScotiaCanada
| | - Stephen L. Bearne
- Department of Biochemistry & Molecular BiologyDalhousie UniversityHalifaxNova ScotiaCanada
- Department of ChemistryDalhousie UniversityHalifaxNova ScotiaCanada
| | - Todd Holyoak
- Department of BiologyUniversity of WaterlooWaterlooOntarioCanada
| |
Collapse
|
25
|
Cheng S, Yang J. A Theoretical Study of Organotin Binding in Aromatase. Int J Mol Sci 2023; 24:ijms24108954. [PMID: 37240300 DOI: 10.3390/ijms24108954] [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: 03/30/2023] [Revised: 05/11/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
The widely used organotin compounds are notorious for their acute toxicity. Experiments revealed that organotin might cause reproductive toxicity by reversibly inhibiting animal aromatase functioning. However, the inhibition mechanism is obscure, especially at the molecular level. Compared to experimental methods, theoretical approaches via computational simulations can help to gain a microscopic view of the mechanism. Here, in an initial attempt to uncover the mechanism, we combined molecular docking and classical molecular dynamics to investigate the binding between organotins and aromatase. The energetics analysis indicated that the van der Waals interaction is the primary driving force of binding the organic tail of organotin and the aromatase center. The hydrogen bond linkage trajectory analysis revealed that water plays a significant role in linking the ligand-water-protein triangle network. As an initial step in studying the mechanism of organotin inhibiting aromatase, this work provides an in-depth understanding of the binding mechanism of organotin. Further, our study will help to develop effective and environmentally friendly methods to treat animals that have already been contaminated by organotin, as well as sustainable solutions for organotin degradation.
Collapse
Affiliation(s)
- Shuming Cheng
- School of Chemical Engineering and Technology, Sun Yat-Sen University, Zhuhai 519082, China
| | - Jing Yang
- School of Chemical Engineering and Technology, Sun Yat-Sen University, Zhuhai 519082, China
| |
Collapse
|
26
|
Ibukun OJ, Gumtya M, Singh S, Shit A, Haldar D. Effect of the spacer on the structure and self-assembly of FF peptide mimetics. SOFT MATTER 2023; 19:3215-3221. [PMID: 37074778 DOI: 10.1039/d3sm00339f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
We have designed and synthesized a series of FF peptide mimetics with conformationally rigid and flexible spacers to study the effect of spacers on their structure and self-assembly. The results help in understanding biomolecular aggregation and provide a strategy to obtain fractal pattern materials. From X-ray single crystal analysis, the m-diaminobenzene appended FF peptide mimetic adopts a duplex structure stabilized by multiple intermolecular hydrogen bonds. There is also a water molecule bridging between two strands of the duplex. Moreover, the duplex is stabilized by three face-to-face, face-to-edge and edge-to-edge π-π interactions. The duplex formation is also supported by mass spectrometry. In higher order packing, the dimeric subunits further self-assembled to form a complex sheet-like structure stabilized by multiple intermolecular hydrogen bonding and π-π stacking interactions. Moreover, the 1,4-butadiene and m-xylylenediamine appended FF peptide mimetics form stimuli-responsive organogels in a wide range of solvents including methanol. The rheology data of FF peptide mimetic gels as a function of angular frequency and oscillatory strain also supported the formation of strong physically crosslinked gels. The FE-SEM images of the xerogels obtained from different organic solvents show that the network morphology of FF peptide mimetics varies depending on the nature of the solvents.
Collapse
Affiliation(s)
- Olamilekan Joseph Ibukun
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, West Bengal, India.
| | - Milan Gumtya
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, West Bengal, India.
| | - Surajit Singh
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, West Bengal, India.
| | - Ananda Shit
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, West Bengal, India.
| | - Debasish Haldar
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, West Bengal, India.
| |
Collapse
|
27
|
Shen Y, Cruz-Cabeza AJ, Azzouz O, Edkins K. Using Prenucleation Aggregation of Caffeine-Benzoic Acid as a Rapid Indication of Co-crystallization from Solutions. Mol Pharm 2023; 20:1942-1950. [PMID: 36942815 DOI: 10.1021/acs.molpharmaceut.2c00829] [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: 03/23/2023]
Abstract
Co-crystal design is a convenient way to remedy the poor biopharmaceutical properties of drugs. Most studies focus on experimental co-crystal screening or computational prediction, but hardly any work has been done toward fast, efficient, and reliable prediction of solution crystallization for co-crystal formation. Here, we study the caffeine-benzoic acid co-crystal system, due to its reported difficulty to crystallize from the solution phase. With this work, we investigate whether there is a link between prenucleation aggregation in solution and co-crystal formation and how to harness this for crystallization prediction. 1H and 13C NMR spectroscopy is used to study the prenucleation interaction between caffeine and benzoic acid in methanol, acetone, and acetonitrile as examples of common solvents. In this system, crystallization from methanol leads to no co-crystallization, from acetone to concomitant crystallization of co-crystal and caffeine, and from acetonitrile to pure co-crystal formation from solution. Strong heteromeric dimers were found to exist in all three solvents. Ternary phase diagrams were defined and a solution-accessible co-crystal region was found for all solvents. For this system, the prenucleation clusters found in solution could be linked to the crystallization of the co-crystal. Crystallization from DMSO did not yield the co-crystal and there were no detectable prenucleation aggregates. NMR spectroscopy to probe dimers in solution can thus be used as a fast, reliable, and promising tool to predict co-crystallization from specific solvents and to screen for suitable solvents for manufacturing and scale-up.
Collapse
Affiliation(s)
- Yichun Shen
- School of Health Sciences, University of Manchester, Manchester M13 9PT, U.K
| | | | - Ossama Azzouz
- School of Health Sciences, University of Manchester, Manchester M13 9PT, U.K
| | - Katharina Edkins
- School of Health Sciences, University of Manchester, Manchester M13 9PT, U.K
| |
Collapse
|
28
|
Hiew SH, Lu Y, Han H, Gonçalves RA, Alfarano SR, Mezzenga R, Parikh AN, Mu Y, Miserez A. Modulation of Mechanical Properties of Short Bioinspired Peptide Materials by Single Amino-Acid Mutations. J Am Chem Soc 2023; 145:3382-3393. [PMID: 36730942 DOI: 10.1021/jacs.2c09853] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The occurrence of modular peptide repeats in load-bearing (structural) proteins is common in nature, with distinctive peptide sequences that often remain conserved across different phylogenetic lineages. These highly conserved peptide sequences endow specific mechanical properties to the material, such as toughness or elasticity. Here, using bioinformatic tools and phylogenetic analysis, we have identified the GX8 peptide with the sequence GLYGGYGX (where X can be any residue) in a wide range of organisms. By simple mutation of the X residue, we demonstrate that GX8 can be self-assembled into various supramolecular structures, exhibiting vastly different physicochemical and viscoelastic properties, from liquid-like coacervate microdroplets to hydrogels to stiff solid materials. A combination of spectroscopic, electron microscopy, mechanical, and molecular dynamics studies is employed to obtain insights into molecular scale interactions driving self-assembly of GX8 peptides, underscoring that π-π stacking and hydrophobic interactions are the drivers of peptide self-assembly, whereas the X residue determines the extent of hydrogen bonding that regulates the macroscopic mechanical response. This study highlights the ability of single amino-acid polymorphism to tune the supramolecular assembly and bulk material properties of GX8 peptides, enabling us to cover a broad range of potential biomedical applications such as hydrogels for tissue engineering or coacervates for drug delivery.
Collapse
Affiliation(s)
- Shu Hui Hiew
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Yang Lu
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Hao Han
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Rui A Gonçalves
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Serena Rosa Alfarano
- Department of Health Sciences and Technology, ETH Zürich, Zürich 8092, Switzerland
| | - Raffaele Mezzenga
- Department of Health Sciences and Technology, ETH Zürich, Zürich 8092, Switzerland
| | - Atul N Parikh
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore.,Departments of Biomedical Engineering and Materials Science & Engineering, University of California, Davis, California 95616, United States
| | - Yuguang Mu
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Ali Miserez
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| |
Collapse
|
29
|
Mondal A, Hazra A, Chattopadhyay MK, Kundu D, Tarai SK, Biswas P, Bhattacharjee A, Mandal S, Banerjee P. Explicating the recognition phenomenon of hazardous nitro-aromatic compound from contaminated environmental and cellular matrices by rationally designed pyridine-functionalized molecular probes. Heliyon 2023; 9:e13620. [PMID: 36873140 PMCID: PMC9975245 DOI: 10.1016/j.heliyon.2023.e13620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/06/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
In the quest of recognizing hazardous nitro-aromatic compounds in water, two pyridine-functionalized Schiff-base chemosensors, DMP ((E)-N-(3,4-dimethoxybenzylidene)(pyridin-2-yl)methanamine)) and MP (4-((E)-((pyridin-2-yl)methylimino)methyl)-2-ethoxyphenol) have been synthesized to detect mutagenic 2,4,6-Trinitrophenol (TNP) in soil, water as well as cellular matrices by producing turn-off emission responses as a combined consequence of PET and RET processes. Several experimental analyses including ESI-MS, FT-IR, photoluminescence, 1H NMR titration, and the theoretical calculations ascertained the formation and sensing efficacies of the chemosensors. The analytical substantiations revealed that structural variation of the chemosensors played a significant role in improving the sensing efficiency, which would certainly be worthwhile in developing small molecular TNP sensors. The present work depicted that the electron density within the MP framework was more than that of DMP due to the intentional incorporation of -OEt and -OH groups. As a result, MP represented a strong interaction mode towards the electron-deficient TNP with a detection limit of 39 μM.
Collapse
Affiliation(s)
- Amita Mondal
- CSIR-Central Mechanical Engineering Research Institute, M. G. Avenue, Durgapur 713209, India.,Department of Chemistry, National Institute of Technology, M. G. Avenue, Durgapur 713209, India
| | - Abhijit Hazra
- CSIR-Central Mechanical Engineering Research Institute, M. G. Avenue, Durgapur 713209, India.,Academy of Scientific and Innovative Research (AcSIR), AcSIR Headquarters CSIR-HRDC Campus, Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad 201002, Uttar Pradesh, India
| | | | - Debojyoti Kundu
- CSIR-Central Mechanical Engineering Research Institute, M. G. Avenue, Durgapur 713209, India.,Academy of Scientific and Innovative Research (AcSIR), AcSIR Headquarters CSIR-HRDC Campus, Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad 201002, Uttar Pradesh, India
| | - Swarup Kumar Tarai
- Department of Chemistry, National Institute of Technology, M. G. Avenue, Durgapur 713209, India
| | - Pritam Biswas
- Department of Biotechnology, National Institute of Technology, M. G. Avenue, Durgapur 713209, India
| | - Ashish Bhattacharjee
- Department of Biotechnology, National Institute of Technology, M. G. Avenue, Durgapur 713209, India
| | - Sukdeb Mandal
- CSIR-Central Mechanical Engineering Research Institute, M. G. Avenue, Durgapur 713209, India.,Academy of Scientific and Innovative Research (AcSIR), AcSIR Headquarters CSIR-HRDC Campus, Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad 201002, Uttar Pradesh, India
| | - Priyabrata Banerjee
- CSIR-Central Mechanical Engineering Research Institute, M. G. Avenue, Durgapur 713209, India.,Academy of Scientific and Innovative Research (AcSIR), AcSIR Headquarters CSIR-HRDC Campus, Postal Staff College Area, Sector 19, Kamla Nehru Nagar, Ghaziabad 201002, Uttar Pradesh, India
| |
Collapse
|
30
|
Hognon C, Bignon E, Monari A, Marazzi M, Garcia-Iriepa C. Revealing the Molecular Interactions between Human ACE2 and the Receptor Binding Domain of the SARS-CoV-2 Wild-Type, Alpha and Delta Variants. Int J Mol Sci 2023; 24:2517. [PMID: 36768842 PMCID: PMC9916449 DOI: 10.3390/ijms24032517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/20/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
After a sudden and first spread of the pandemic caused by the novel SARS-CoV-2 (Severe Acute Respiratory Syndrome-Coronavirus 2) wild-type strain, mutants have emerged which have been associated with increased infectivity, inducing surges in the contagions. The first of the so-called variants of concerns, was firstly isolated in the United Kingdom and later renamed Alpha variant. Afterwards, in the middle of 2021, a new variant appeared called Delta. The latter is characterized by the presence of point mutations in the Spike protein of SARS-CoV-2, especially in the Receptor Binding Domain (RBD). When in its active conformation, the RBD can interact with the human receptor Angiotensin-Converting Enzyme 2 (ACE2) to allow the entry of the virions into cells. In this contribution, by using extended all-atom molecular dynamic simulations, complemented with machine learning post-processing, we analyze the changes in the molecular interaction network induced by these different strains in comparison with the wild-type. On one hand, although relevant variations are evidenced, only limited changes in the global stability indicators and in the flexibility profiles have been observed. On the other hand, key differences were obtained by tracking hydrophilic and hydrophobic molecular interactions, concerning both positioning at the ACE2/RBD interface and formation/disruption dynamic behavior.
Collapse
Affiliation(s)
- Cécilia Hognon
- Departamento de Química Analítica, Química Física e Ingeniería Química, Universidad de Alcalá, Ctra. Madrid-Barcelona, Km 33,600, 28871 Alcalá de Henares, Madrid, Spain
| | - Emmanuelle Bignon
- UMR 7019 LPCT, Université de Lorraine and CNRS, F-5400 Nancy, France
| | - Antonio Monari
- ITODYS, Université Paris Cité and CNRS, F-75006 Paris, France
| | - Marco Marazzi
- Departamento de Química Analítica, Química Física e Ingeniería Química, Universidad de Alcalá, Ctra. Madrid-Barcelona, Km 33,600, 28871 Alcalá de Henares, Madrid, Spain
- Instituto de Investigación Química “Andrés M. del Río” (IQAR), Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain
| | - Cristina Garcia-Iriepa
- Departamento de Química Analítica, Química Física e Ingeniería Química, Universidad de Alcalá, Ctra. Madrid-Barcelona, Km 33,600, 28871 Alcalá de Henares, Madrid, Spain
- Instituto de Investigación Química “Andrés M. del Río” (IQAR), Universidad de Alcalá, 28871 Alcalá de Henares, Madrid, Spain
| |
Collapse
|
31
|
Reza Masoodi H, Sadat Pourhosseini R, Bagheri S. The role of nature of aromatic ring on cooperativity between π–π stacking and ion–π interactions: a computational study. COMPUT THEOR CHEM 2023. [DOI: 10.1016/j.comptc.2023.114022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
32
|
Polypyridyl coordinated rhenium(I) tricarbonyl complexes as model devices for cancer diagnosis and treatment. Polyhedron 2022. [DOI: 10.1016/j.poly.2022.116178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
33
|
Patarroyo ME, Bermudez A, Alba MP, Patarroyo MA, Suarez C, Aza-Conde J, Moreno-Vranich A, Vanegas M. Stereo electronic principles for selecting fully-protective, chemically-synthesised malaria vaccines. Front Immunol 2022; 13:926680. [DOI: 10.3389/fimmu.2022.926680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
Major histocompatibility class II molecule-peptide-T-cell receptor (MHCII-p-TCR) complex-mediated antigen presentation for a minimal subunit-based, multi-epitope, multistage, chemically-synthesised antimalarial vaccine is essential for inducing an appropriate immune response. Deep understanding of this MHCII-p-TCR complex’s stereo-electronic characteristics is fundamental for vaccine development. This review encapsulates the main principles for achieving such epitopes’ perfect fit into MHC-II human (HLADRβ̞1*) or Aotus (Aona DR) molecules. The enormous relevance of several amino acids’ physico-chemical characteristics is analysed in-depth, as is data regarding a 26.5 ± 2.5Å distance between the farthest atoms fitting into HLA-DRβ1* structures’ Pockets 1 to 9, the role of polyproline II-like (PPIIL) structures having their O and N backbone atoms orientated for establishing H-bonds with specific HLA-DRβ1*-peptide binding region (PBR) residues. The importance of residues having specific charge and orientation towards the TCR for inducing appropriate immune activation, amino acids’ role and that of structures interfering with PPIIL formation and other principles are demonstrated which have to be taken into account when designing immune, protection-inducing peptide structures (IMPIPS) against diseases scourging humankind, malaria being one of them.
Collapse
|
34
|
Nulek T, Klaysri R, Cedeno R, Nalaoh P, Bureekaew S, Promarak V, Flood AE. Separation of Etiracetam Enantiomers Using Enantiospecific Cocrystallization with 2-Chloromandelic Acid. ACS OMEGA 2022; 7:19465-19473. [PMID: 35721919 PMCID: PMC9202017 DOI: 10.1021/acsomega.2c01165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 05/16/2022] [Indexed: 06/15/2023]
Abstract
Chirality plays an important role in the pharmaceutical industry since the two enantiomers of a drug molecule usually display significantly different bioactivities, and hence, most products are produced as pure enantiomers. However, many drug precursors are synthesized as racemates, and hence, enantioseparation has become a significant process in the industry. Cocrystallization is one of the attractive crystallization approaches to obtain the desired enantiomer from racemic compounds. In this work, we propose a chiral resolution route for an antiepileptic drug, S-etiracetam (S-ETI), via enantiospecific cocrystallization with S-2-chloro-S-mandelic acid (CLMA) as a coformer. The experiments indicate that the system is highly enantiospecific; S-2CLMA cocrystallizes only with S-ETI but not with R-ETI or RS-ETI. Therefore, the chiral purification of S-ETI can be achieved efficiently with a 69.1% yield and close to 100% enantiopurity from the racemic solution. Additionally, structural simulations of the S-ETI:S-2CLMA cocrystal reveal that the cocrystal structure has higher thermodynamic stability than that of R-ETI:S-2CLMA by about 5.5 kcal/mol (per cocrystal formula unit), which helps to confirm the favorability of the enantiospecification in this system.
Collapse
Affiliation(s)
- Thitapond Nulek
- Department
of Chemical and Biomolecular Engineering, School of Energy Science
and Engineering, Vidyasirimedhi Institute
of Science and Technology, 555 Moo 1, Payupnai, Wang Chan, 21210 Rayong, Thailand
| | - Rachan Klaysri
- Department
of Chemical and Biomolecular Engineering, School of Energy Science
and Engineering, Vidyasirimedhi Institute
of Science and Technology, 555 Moo 1, Payupnai, Wang Chan, 21210 Rayong, Thailand
| | - Ruel Cedeno
- Department
of Chemical and Biomolecular Engineering, School of Energy Science
and Engineering, Vidyasirimedhi Institute
of Science and Technology, 555 Moo 1, Payupnai, Wang Chan, 21210 Rayong, Thailand
| | - Phattananawee Nalaoh
- Department
of Materials Science and Engineering, School of Molecular Science
and Engineering, Vidyasirimedhi Institute
of Science and Technology, 555 Moo 1, Payupnai, Wang Chan, 21210 Rayong, Thailand
| | - Sareeya Bureekaew
- Department
of Chemical and Biomolecular Engineering, School of Energy Science
and Engineering, Vidyasirimedhi Institute
of Science and Technology, 555 Moo 1, Payupnai, Wang Chan, 21210 Rayong, Thailand
| | - Vinich Promarak
- Department
of Materials Science and Engineering, School of Molecular Science
and Engineering, Vidyasirimedhi Institute
of Science and Technology, 555 Moo 1, Payupnai, Wang Chan, 21210 Rayong, Thailand
| | - Adrian E. Flood
- Department
of Chemical and Biomolecular Engineering, School of Energy Science
and Engineering, Vidyasirimedhi Institute
of Science and Technology, 555 Moo 1, Payupnai, Wang Chan, 21210 Rayong, Thailand
| |
Collapse
|
35
|
Aguayo-Ortiz R, Dominguez L. Unveiling the Possible Oryzalin-Binding Site in the α-Tubulin of Toxoplasma gondii. ACS OMEGA 2022; 7:18434-18442. [PMID: 35694483 PMCID: PMC9178734 DOI: 10.1021/acsomega.2c00729] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/11/2022] [Indexed: 06/09/2023]
Abstract
Dinitroaniline derivatives have been widely used as herbicidal agents to control weeds and grass. Previous studies demonstrated that these compounds also exhibit good antiparasitic activity against some protozoan parasites. Oryzalin (ORY), a representative dinitroaniline derivative, exerts its antiprotozoal activity against Toxoplasma gondii by inhibiting the microtubule polymerization process. Moreover, the identification of ORY-resistant T. gondii lines obtained by chemical mutagenesis confirmed that this compound binds selectively to α-tubulin. Based on experimental information reported so far and a multiple sequence analysis carried out in this work, we propose that the pironetin (PIR) site is the potential ORY-binding site. Therefore, we employed state-of-the-art computational approaches to characterize the interaction profile of ORY at the proposed site in the α-tubulin of T. gondii. An exhaustive search for other possible binding sites was performed using the Wrap "N" Shake method, which showed that ORY exhibits highest stability and affinity for the PIR site. Moreover, our molecular dynamics simulations revealed that the dipropylamine substituent of ORY interacts with a hydrophobic pocket, while the sulfonamide group formed hydrogen bonds with water molecules at the site entrance. Overall, our results suggest that ORY binds to the PIR site on the α-tubulin of the protozoan parasite T. gondii. This information will be very useful for designing less toxic and more potent antiprotozoal agents.
Collapse
Affiliation(s)
- Rodrigo Aguayo-Ortiz
- Departamento
de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Laura Dominguez
- Departamento
de Fisicoquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
36
|
Leśniak RK, Nichols RJ, Schonemann M, Zhao J, Gajera CR, Lam G, Nguyen KC, Langston JW, Smith M, Montine TJ. Discovery of 1 H-Pyrazole Biaryl Sulfonamides as Novel G2019S-LRRK2 Kinase Inhibitors. ACS Med Chem Lett 2022; 13:981-988. [DOI: 10.1021/acsmedchemlett.2c00116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/18/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Robert K. Leśniak
- Medicinal Chemistry Knowledge Center, Sarafan ChEM-H, Stanford University, Stanford, California 94305, United States
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, California 94305, United States
| | - R. Jeremy Nichols
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, California 94305, United States
| | - Marcus Schonemann
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, California 94305, United States
| | - Jing Zhao
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, California 94305, United States
| | - Chandresh R. Gajera
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, California 94305, United States
| | - Grace Lam
- Departments of Medicine and Microbiology and Immunology, Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Khanh C. Nguyen
- Departments of Medicine and Microbiology and Immunology, Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - J. William Langston
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, California 94305, United States
- Department of Neurology and Neuroscience, Stanford University, 300 Pasteur Drive, Stanford, California 94304, United States
| | - Mark Smith
- Medicinal Chemistry Knowledge Center, Sarafan ChEM-H, Stanford University, Stanford, California 94305, United States
| | - Thomas J. Montine
- Department of Pathology, Stanford University, 300 Pasteur Drive, Stanford, California 94305, United States
| |
Collapse
|
37
|
Wang Y, Fu M, Zhang X, Jin D, Zhu S, Wang Y, Wu Z, Bao J, Cheng X, Yang L, Xie L. Cubic Nanogrids for Counterbalance Contradiction among Reorganization Energy, Strain Energy, and Wide Bandgap. J Phys Chem Lett 2022; 13:4297-4308. [PMID: 35532545 DOI: 10.1021/acs.jpclett.2c00827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Molecular cross-scale gridization and polygridization of organic π-backbones make it possible to install 0/1/2/3-dimensional organic wide-bandgap semiconductors (OWBGSs) with potentially ZnO-like fascinating multifunctionality such as optoelectronic and piezoelectronic features. However, gridization effects are limited to uncover, because the establishment of gridochemistry still requires a long time, which offers a chance to understand the effects with a theoretical method, together with data statistics and machine learning. Herein, we demonstrate a state-of-the-art 3D cubic nanogridon with a size of ∼2 × 2 × 1.5 nm3 to examine its multigridization of π-segments on the bandgap, molecular strain energy (MSE), as well as reorganization energy (ROE). A cubic gridon (CG) consists of a four-armed bifluorene skeleton and a thiophene-containing fused arene plane with the Csp3 spiro-linkage, which can be deinstalled into face-on or edge-on monogrids. As a result, multigridization does not significantly reduce bandgaps (Eg ≥ 4.03 eV), while the MSE increases gradually from 4.72 to 23.83 kcal/mol. Very importantly, the ROE of a CG exhibits an extreme reduction down to ∼28 meV (λ+) that is near the thermal fluctuation energy (∼26 meV). Our multigridization results break through the limitation of the basic positively proportional relationship between reorganization energies and bandgaps in organic semiconductors. Furthermore, multigridization makes it possible to keep the ROE small under the condition of a high MSE in OWBGS that will guide the cross-scale design of multifunctional OWBGSs with both inorganics' optoelectronic performance and organics' mechanical flexibility.
Collapse
Affiliation(s)
- Yongxia Wang
- Center for Molecular Systems & Organic Devices (CMSOD), Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Mingyang Fu
- Center for Molecular Systems & Organic Devices (CMSOD), Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Xiaofei Zhang
- Institute of Agricultural Remote Sensing and Information, Heilongjiang Academy of Agricultural Sciences, Harbin 150086, China
| | - Dong Jin
- Center for Molecular Systems & Organic Devices (CMSOD), Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Shiyuan Zhu
- Center for Molecular Systems & Organic Devices (CMSOD), Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Yucong Wang
- Center for Molecular Systems & Organic Devices (CMSOD), Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Zhenyu Wu
- School of Internet of Things, Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Jianmin Bao
- School of Internet of Things, Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Xiaogang Cheng
- School of Communications and Information Engineering, Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Lei Yang
- Center for Molecular Systems & Organic Devices (CMSOD), Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Linghai Xie
- Center for Molecular Systems & Organic Devices (CMSOD), Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| |
Collapse
|
38
|
Lu H, Fang L, Wang X, Wu D, Liu C, Liu X, Wang J, Gao Y, Min W. Structure-Activity Relationship of Pine Nut-Derived Peptides and Their Protective Effect on Nerve-Cell Mitochondria. Foods 2022; 11:foods11101428. [PMID: 35626998 PMCID: PMC9140850 DOI: 10.3390/foods11101428] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/14/2022] [Accepted: 05/14/2022] [Indexed: 12/02/2022] Open
Abstract
This study aimed to investigate the structure-activity relationship of the pine nut antioxidant peptide WYPGK and its derivative peptides, and to evaluate the protective effect of the latter on oxidative damage to mitochondrial structure and function in PC12 cells. Molecular docking revealed the derivative peptides WYFGK and WYSGK to have higher affinity to the active region of sirtuin 3 (SIRT3) (−6.08 kcal/mol and −5.87 kcal/mol, respectively), hence indicating that they are promising SIRT3 inducers and antioxidant factors. The derivative peptide WYSGK presented the highest ORAC value (5457.70 µmol TE/g), ABTS scavenging activity (70.05%), and Fe2+-chelating activity (81.70%), followed by WYPGK and WYFGK. Circular dichroism and nuclear magnetic resonance data suggested that the presence of 3-Ser in WYSGK increased its β-sheet content, and that the active hydrogen atoms produced chemical shifts. In H2O2-induced PC12 cells, WYSGK substantially reduced ROS and MDA levels, and increased ATP levels. Transmission electron microscopy and Seahorse Analyze assay proved the peptide WYSGK to significantly alleviate mitochondrial damage and respiratory dysfunction (p < 0.05), thereby implying that a study of structure-activity relationships of the peptides can possibly be an effective approach for the development of functional factors.
Collapse
Affiliation(s)
- Hongyan Lu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (H.L.); (L.F.); (X.W.); (D.W.); (C.L.); (X.L.); (J.W.); (Y.G.)
- National Engineering Laboratory of Wheat and Corn Deep Processing, Changchun 130118, China
| | - Li Fang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (H.L.); (L.F.); (X.W.); (D.W.); (C.L.); (X.L.); (J.W.); (Y.G.)
- National Engineering Laboratory of Wheat and Corn Deep Processing, Changchun 130118, China
| | - Xiyan Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (H.L.); (L.F.); (X.W.); (D.W.); (C.L.); (X.L.); (J.W.); (Y.G.)
- National Engineering Laboratory of Wheat and Corn Deep Processing, Changchun 130118, China
| | - Dan Wu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (H.L.); (L.F.); (X.W.); (D.W.); (C.L.); (X.L.); (J.W.); (Y.G.)
- National Engineering Laboratory of Wheat and Corn Deep Processing, Changchun 130118, China
| | - Chunlei Liu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (H.L.); (L.F.); (X.W.); (D.W.); (C.L.); (X.L.); (J.W.); (Y.G.)
- National Engineering Laboratory of Wheat and Corn Deep Processing, Changchun 130118, China
| | - Xiaoting Liu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (H.L.); (L.F.); (X.W.); (D.W.); (C.L.); (X.L.); (J.W.); (Y.G.)
- National Engineering Laboratory of Wheat and Corn Deep Processing, Changchun 130118, China
| | - Ji Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (H.L.); (L.F.); (X.W.); (D.W.); (C.L.); (X.L.); (J.W.); (Y.G.)
- National Engineering Laboratory of Wheat and Corn Deep Processing, Changchun 130118, China
| | - Yawen Gao
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (H.L.); (L.F.); (X.W.); (D.W.); (C.L.); (X.L.); (J.W.); (Y.G.)
- National Engineering Laboratory of Wheat and Corn Deep Processing, Changchun 130118, China
| | - Weihong Min
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China; (H.L.); (L.F.); (X.W.); (D.W.); (C.L.); (X.L.); (J.W.); (Y.G.)
- National Engineering Laboratory of Wheat and Corn Deep Processing, Changchun 130118, China
- Correspondence: ; Tel.: +86-139-4491-9697; Fax: +86-431-8451-7235
| |
Collapse
|
39
|
Beyett TS, To C, Heppner DE, Rana JK, Schmoker AM, Jang J, De Clercq DJH, Gomez G, Scott DA, Gray NS, Jänne PA, Eck MJ. Molecular basis for cooperative binding and synergy of ATP-site and allosteric EGFR inhibitors. Nat Commun 2022; 13:2530. [PMID: 35534503 PMCID: PMC9085736 DOI: 10.1038/s41467-022-30258-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/19/2022] [Indexed: 12/22/2022] Open
Abstract
Lung cancer is frequently caused by activating mutations in the epidermal growth factor receptor (EGFR). Allosteric EGFR inhibitors offer promise as the next generation of therapeutics, as they are unaffected by common ATP-site resistance mutations and synergize with the drug osimertinib. Here, we examine combinations of ATP-competitive and allosteric inhibitors to better understand the molecular basis for synergy. We identify a subset of irreversible EGFR inhibitors that display positive binding cooperativity and synergy with the allosteric inhibitor JBJ-04-125-02 in several EGFR variants. Structural analysis of these complexes reveals conformational changes occur mainly in the phosphate-binding loop (P-loop). Mutation of F723 in the P-loop reduces cooperative binding and synergy, supporting a mechanism in which F723-mediated contacts between the P-loop and the allosteric inhibitor are critical for synergy. These structural and mechanistic insights will aid in the identification and development of additional inhibitor combinations with potential clinical value. Acquired drug resistance is common during chemotherapy. Here, the authors describe the structural basis and molecular mechanism by which allosteric and clinically approved, ATP-competitive inhibitors of EGFR synergize to overcome resistance in lung cancer.
Collapse
|
40
|
Pan C, Chen L, Zhang X, Zhang D, Song Q, Peng J, Li Q. Molecular insight into the
π‐stacking
interactions of human ovarian cancer
PARP
‐1 with its small‐molecule inhibitors and rational design of aromatic amino acid‐rich peptides to target
PARP
‐1 based on the
π‐stacking
network. J CHIN CHEM SOC-TAIP 2022. [DOI: 10.1002/jccs.202200004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Chunxia Pan
- Department of Gynecological Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan China
| | - Lei Chen
- Department of Gynecological Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan China
| | - Xinxin Zhang
- Department of Gynecological Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan China
| | - Depu Zhang
- Department of Gynecological Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan China
| | - Quqing Song
- Department of Gynecological Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan China
| | - Jingwei Peng
- Department of Gynecological Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan China
| | - Qingshui Li
- Department of Gynecological Oncology, Shandong Cancer Hospital and Institute Shandong First Medical University and Shandong Academy of Medical Sciences Jinan China
| |
Collapse
|
41
|
Gu J, Wang X, Zhao W, Zhuang R, Zhang C, Zhang X, Cai Y, Yuan W, Luan B, Dong B, Liu H. Synthesis of Half-Titanocene Complexes Containing π,π-Stacked Aryloxide Ligands, and Their Use as Catalysts for Ethylene (Co)polymerizations. Polymers (Basel) 2022; 14:polym14071427. [PMID: 35406299 PMCID: PMC9002825 DOI: 10.3390/polym14071427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/22/2022] [Accepted: 03/28/2022] [Indexed: 01/27/2023] Open
Abstract
A family of half-titanocene complexes bearing π,π-stacked aryloxide ligands and their catalytic performances towards ethylene homo-/co- polymerizations were disclosed herein. All the complexes were well characterized, and the intermolecular π,π-stacking interactions could be clearly identified from single crystal X-ray analysis, in which a stronger interaction could be reflected for aryloxides bearing bigger π-systems, e.g., pyrenoxide. Due to the formation of such interactions, these complexes were able to highly catalyze the ethylene homopolymerizations and copolymerization with 1-hexene comonomer, even without any additiveson the aryloxide group, which showed striking contrast to other half-titanocene analogues, implying the positive influence of π,π-stacking interaction in enhancing the catalytic performances of the corresponding catalysts. Moreover, it was found that addition of external pyrene molecules was capable of boosting the catalytic efficiency significantly, due to the formation of a stronger π,π-stacking interaction between the complexes and pyrene molecules.
Collapse
Affiliation(s)
- Jin Gu
- Shandong Provincial Key Laboratory of Olefin Catalysis and Polymerization, Chambroad Chemical Industry Research Institute Co., Ltd., Qingdao 266042, China; (J.G.); (R.Z.); (Y.C.); (W.Y.); (B.L.)
| | - Xiaohua Wang
- Key Laboratory of Rubber-Plastics, Ministry of Education/Shandong Provincial Key Laboratory of Rubber-Plastics, Qingdao University of Science & Technology, Qingdao 266042, China; (X.W.); (W.Z.); (C.Z.); (X.Z.)
| | - Wenpeng Zhao
- Key Laboratory of Rubber-Plastics, Ministry of Education/Shandong Provincial Key Laboratory of Rubber-Plastics, Qingdao University of Science & Technology, Qingdao 266042, China; (X.W.); (W.Z.); (C.Z.); (X.Z.)
| | - Rui Zhuang
- Shandong Provincial Key Laboratory of Olefin Catalysis and Polymerization, Chambroad Chemical Industry Research Institute Co., Ltd., Qingdao 266042, China; (J.G.); (R.Z.); (Y.C.); (W.Y.); (B.L.)
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Chunyu Zhang
- Key Laboratory of Rubber-Plastics, Ministry of Education/Shandong Provincial Key Laboratory of Rubber-Plastics, Qingdao University of Science & Technology, Qingdao 266042, China; (X.W.); (W.Z.); (C.Z.); (X.Z.)
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Xuequan Zhang
- Key Laboratory of Rubber-Plastics, Ministry of Education/Shandong Provincial Key Laboratory of Rubber-Plastics, Qingdao University of Science & Technology, Qingdao 266042, China; (X.W.); (W.Z.); (C.Z.); (X.Z.)
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Yinghui Cai
- Shandong Provincial Key Laboratory of Olefin Catalysis and Polymerization, Chambroad Chemical Industry Research Institute Co., Ltd., Qingdao 266042, China; (J.G.); (R.Z.); (Y.C.); (W.Y.); (B.L.)
| | - Wenbo Yuan
- Shandong Provincial Key Laboratory of Olefin Catalysis and Polymerization, Chambroad Chemical Industry Research Institute Co., Ltd., Qingdao 266042, China; (J.G.); (R.Z.); (Y.C.); (W.Y.); (B.L.)
| | - Bo Luan
- Shandong Provincial Key Laboratory of Olefin Catalysis and Polymerization, Chambroad Chemical Industry Research Institute Co., Ltd., Qingdao 266042, China; (J.G.); (R.Z.); (Y.C.); (W.Y.); (B.L.)
| | - Bo Dong
- Shandong Provincial Key Laboratory of Olefin Catalysis and Polymerization, Chambroad Chemical Industry Research Institute Co., Ltd., Qingdao 266042, China; (J.G.); (R.Z.); (Y.C.); (W.Y.); (B.L.)
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- Correspondence: (B.D.); (H.L.)
| | - Heng Liu
- Key Laboratory of Rubber-Plastics, Ministry of Education/Shandong Provincial Key Laboratory of Rubber-Plastics, Qingdao University of Science & Technology, Qingdao 266042, China; (X.W.); (W.Z.); (C.Z.); (X.Z.)
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- Correspondence: (B.D.); (H.L.)
| |
Collapse
|
42
|
Lee JA, Kwon YW, Kim HR, Shin N, Son HJ, Cheong CS, Kim DJ, Hwang O. A Novel Pyrazolo[3,4- d]pyrimidine Induces Heme Oxygenase-1 and Exerts Anti-Inflammatory and Neuroprotective Effects. Mol Cells 2022; 45:134-147. [PMID: 34887364 PMCID: PMC8926863 DOI: 10.14348/molcells.2021.0074] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 09/14/2021] [Accepted: 10/15/2021] [Indexed: 11/27/2022] Open
Abstract
The anti-oxidant enzyme heme oxygenase-1 (HO-1) is known to exert anti-inflammatory effects. From a library of pyrazolo[3,4-d]pyrimidines, we identified a novel compound KKC080096 that upregulated HO-1 at the mRNA and protein levels in microglial BV-2 cells. KKC080096 exhibited anti-inflammatory effects via suppressing nitric oxide, interleukin-1β (IL-1β), and iNOS production in lipopolysaccharide (LPS)-challenged cells. It inhibited the phosphorylation of IKK and MAP kinases (p38, JNK, ERK), which trigger inflammatory signaling, and whose activities are inhibited by HO-1. Further, KKC080096 upregulated anti-inflammatory marker (Arg1, YM1, CD206, IL-10, transforming growth factor-β [TGF-β]) expression. In 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mice, KKC080096 lowered microglial activation, protected the nigral dopaminergic neurons, and nigral damage-associated motor deficits. Next, we elucidated the mechanisms by which KKC080096 upregulated HO-1. KKC080096 induced the phosphorylation of AMPK and its known upstream kinases LKB1 and CaMKKbeta, and pharmacological inhibition of AMPK activity reduced the effects of KKC080096 on HO-1 expression and LPS-induced NO generation, suggesting that KKC080096-induced HO-1 upregulation involves LKB1/AMPK and CaMKKbeta/AMPK pathway activation. Further, KKC080096 caused an increase in cellular Nrf2 level, bound to Keap1 (Nrf2 inhibitor protein) with high affinity, and blocked Keap1-Nrf2 interaction. This Nrf2 activation resulted in concurrent induction of HO-1 and other Nrf2-targeted antioxidant enzymes in BV-2 and in dopaminergic CATH.a cells. These results indicate that KKC080096 is a potential therapeutic for oxidative stress- and inflammation-related neurodegenerative disorders such as Parkinson's disease.
Collapse
Affiliation(s)
- Ji Ae Lee
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Young-Won Kwon
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Hye Ri Kim
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Nari Shin
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Hyo Jin Son
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Chan Seong Cheong
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Dong Jin Kim
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - Onyou Hwang
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul 05505, Korea
| |
Collapse
|
43
|
Wojciechowska A, de Graaf C, Rojek T, Jerzykiewicz M, Malik M, Gągor A, Duczmal M. A Rare Diiodo−L−tyrosine Copper(II) Complexes – crystal and molecular structure of materials stabilized by weak interactions. Polyhedron 2022. [DOI: 10.1016/j.poly.2022.115780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
44
|
Kiss M, Timári I, Barna T, Mészáros Z, Slámová K, Bojarová P, Křen V, Hayes JM, Somsák L. 2-Acetamido-2-deoxy-d-glucono-1,5-lactone Sulfonylhydrazones: Synthesis and Evaluation as Inhibitors of Human OGA and HexB Enzymes. Int J Mol Sci 2022; 23:ijms23031037. [PMID: 35162960 PMCID: PMC8834866 DOI: 10.3390/ijms23031037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/06/2022] [Accepted: 01/15/2022] [Indexed: 01/27/2023] Open
Abstract
Inhibition of the human O-linked β-N-acetylglucosaminidase (hOGA, GH84) enzyme is pharmacologically relevant in several diseases such as neurodegenerative and cardiovascular disorders, type 2 diabetes, and cancer. Human lysosomal hexosaminidases (hHexA and hHexB, GH20) are mechanistically related enzymes; therefore, selective inhibition of these enzymes is crucial in terms of potential applications. In order to extend the structure–activity relationships of OGA inhibitors, a series of 2-acetamido-2-deoxy-d-glucono-1,5-lactone sulfonylhydrazones was prepared from d-glucosamine. The synthetic sequence involved condensation of N-acetyl-3,4,6-tri-O-acetyl-d-glucosamine with arenesulfonylhydrazines, followed by MnO2 oxidation to the corresponding glucono-1,5-lactone sulfonylhydrazones. Removal of the O-acetyl protecting groups by NH3/MeOH furnished the test compounds. Evaluation of these compounds by enzyme kinetic methods against hOGA and hHexB revealed potent nanomolar competitive inhibition of both enzymes, with no significant selectivity towards either. The most efficient inhibitor of hOGA was 2-acetamido-2-deoxy-d-glucono-1,5-lactone 1-naphthalenesulfonylhydrazone (5f, Ki = 27 nM). This compound had a Ki of 6.8 nM towards hHexB. To assess the binding mode of these inhibitors to hOGA, computational studies (Prime protein–ligand refinement and QM/MM optimizations) were performed, which suggested the binding preference of the glucono-1,5-lactone sulfonylhydrazones in an s-cis conformation for all test compounds.
Collapse
Affiliation(s)
- Mariann Kiss
- Department of Organic Chemistry, University of Debrecen, POB 400, H-4002 Debrecen, Hungary; (M.K.); (I.T.)
| | - István Timári
- Department of Organic Chemistry, University of Debrecen, POB 400, H-4002 Debrecen, Hungary; (M.K.); (I.T.)
| | - Teréz Barna
- Department of Genetics and Applied Microbiology, University of Debrecen, POB 400, H-4002 Debrecen, Hungary;
| | - Zuzana Mészáros
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220 Praha 4, Czech Republic; (Z.M.); (K.S.); (P.B.); (V.K.)
- Faculty of Food and Biochemical Technology, University of Chemistry and Technology Prague, Technická 1903/3, CZ-16628 Praha 6, Czech Republic
| | - Kristýna Slámová
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220 Praha 4, Czech Republic; (Z.M.); (K.S.); (P.B.); (V.K.)
| | - Pavla Bojarová
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220 Praha 4, Czech Republic; (Z.M.); (K.S.); (P.B.); (V.K.)
| | - Vladimír Křen
- Laboratory of Biotransformation, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, CZ-14220 Praha 4, Czech Republic; (Z.M.); (K.S.); (P.B.); (V.K.)
| | - Joseph M. Hayes
- School of Pharmacy & Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK;
| | - László Somsák
- Department of Organic Chemistry, University of Debrecen, POB 400, H-4002 Debrecen, Hungary; (M.K.); (I.T.)
- Correspondence:
| |
Collapse
|
45
|
Hiiuk VM, Shylin SI, Barakhtii DD, Korytko DM, Kotsyubynsky VO, Rotaru A, Shova S, Gural'skiy IA. Two-Step Spin Crossover in Hofmann-Type Coordination Polymers [Fe(2-phenylpyrazine) 2{M(CN) 2} 2] (M = Ag, Au). Inorg Chem 2022; 61:2093-2104. [PMID: 35029111 DOI: 10.1021/acs.inorgchem.1c03302] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Two 2D Hofmann-type complexes of the composition [Fe(Phpz)2{M(CN)2}2] (where Phpz = 2-phenylpyrazine; M = Ag, Au) have been synthesized, and their spin-crossover (SCO) behavior has been thoroughly characterized. Single-crystal X-ray analysis reveals that these complexes contain a crystallographically unique Fe(II) center surrounded by two axial Phpz ligands and four equatorial cyanide [M(CN)2]- bridges. It is shown that, using of a ligand with two aromatic rings, an advanced system of weak supramolecular interactions (metal-metal, C-H···M, and π···π stacking contacts) is realized. This ensures additional stabilization of the structures and the absence of solvent-accessible voids due to dense packing. Both complexes are characterized by a highly reproducible two-step SCO behavior, as revealed by different techniques (superconducting quantum interference device magnetometry, optical microscopy, etc.). Research shows the exceptional role of the presence of various supramolecular interactions in the structure and the influence of the bulky substituent in the ligand on SCO behavior. Moreover, the perspective of substituted pyrazines for the design of new switchable materials is supported by this work.
Collapse
Affiliation(s)
- Volodymyr M Hiiuk
- Department of Chemistry, Taras Shevchenko National University of Kyiv, 64 Volodymyrska Street, 01601 Kyiv, Ukraine.,Faculty of Natural Sciences, National University of Kyiv-Mohyla Academy, 2 Skovorody Street, 04070 Kyiv, Ukraine
| | - Sergii I Shylin
- Ångström Laboratory, Department of Chemistry, Uppsala University, 75120 Uppsala, Sweden
| | - Diana D Barakhtii
- Department of Chemistry, Taras Shevchenko National University of Kyiv, 64 Volodymyrska Street, 01601 Kyiv, Ukraine
| | - Dmytro M Korytko
- Department of Chemistry, Taras Shevchenko National University of Kyiv, 64 Volodymyrska Street, 01601 Kyiv, Ukraine
| | - Volodymyr O Kotsyubynsky
- Department of Material Science and New Technology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenko Street, 76018 Ivano-Frankivsk, Ukraine
| | - Aurelian Rotaru
- Faculty of Electrical Engineering and Computer Science & Research Center MANSiD, Stefan cel Mare University, 13 Universitatii Street, 720229 Suceava, Romania
| | - Sergiu Shova
- Ningbo University of Technology, No. 201, Fenghua Road, Ningbo City, Zhejiang 315211, China.,"Petru Poni" Institute of Macromolecular Chemistry, 41A Aleea Gr. Ghica Voda, 700487 Iasi, Romania
| | - Il'ya A Gural'skiy
- Department of Chemistry, Taras Shevchenko National University of Kyiv, 64 Volodymyrska Street, 01601 Kyiv, Ukraine
| |
Collapse
|
46
|
Roy Chowdhury S, Haldar D. A gama-turn mimetic for selective sensing of Cu(II) and combinatorial multiple logic gate. CrystEngComm 2022. [DOI: 10.1039/d2ce00462c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We have designed and synthesized a gama-turn mimetic using fenamic acid and α-aminoisobutyricacid (Aib), the conformation and optoelectronic properties of which can be changed by appropriate external stimuli. From single-crystal...
Collapse
|
47
|
Xiao R, Yu G, Xu BB, Wang N, Liu X. Fiber Surface/Interfacial Engineering on Wearable Electronics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2102903. [PMID: 34418304 DOI: 10.1002/smll.202102903] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/29/2021] [Indexed: 06/13/2023]
Abstract
Surface/interfacial engineering is an essential technique to explore the fiber materials properties and fulfil new functionalities. An extensive scope of current physical and chemical treating methods is reviewed here together with a variety of real-world applications. Moreover, a new surface/interface engineering approach is also introduced: self-assembly via π-π stacking, which has great potential for the surface modification of fiber materials due to its nondestructive working principle. A new fiber family member, metal-oxide framework (MOF) fiber shows promising candidacy for fiber based wearable electronics. The understanding of surface/interfacial engineering techniques on fiber materials is advanced here and it is expected to guide the rational design of future fiber based wearable electronics.
Collapse
Affiliation(s)
- Ruimin Xiao
- Department of Materials, Faculty of Science and Engineering, University of Manchester, Oxford Rd., Manchester, M13 9PL, UK
| | - Guiqin Yu
- College of Chemistry and Chemical Engineering, Lanzhou University, 222 Tianshui Southern Road, Lanzhou, Gansu, 730000, China
| | - Ben Bin Xu
- Mechanical and Construction Engineering, Faculty of Engineering and Environment, Northumbria University, Newcastle upon Tyne, NE1 8ST, UK
| | - Nan Wang
- The Nanoscience Centre, University of Cambridge, Cambridge, CB3 0FF, UK
| | - Xuqing Liu
- Department of Materials, Faculty of Science and Engineering, University of Manchester, Oxford Rd., Manchester, M13 9PL, UK
| |
Collapse
|
48
|
Lai YY, Li D, Chang SW. Computational insights into the substrate recognition mechanism of cartilage extracellular matrix degradation. Comput Struct Biotechnol J 2021; 19:5535-5545. [PMID: 34712398 PMCID: PMC8526910 DOI: 10.1016/j.csbj.2021.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/01/2021] [Accepted: 10/01/2021] [Indexed: 11/23/2022] Open
Abstract
Articular cartilage is connective tissue that forms a slippery load-bearing joint surface between bones. With outstanding mechanical properties, it plays an essential role in cushioning impact and protecting the ends of bones. Abnormal mechanical stimulation, such as repetitive overloading or chondral injury, induces excessive cartilage extracellular matrix (ECM) degradation, leading to osteoarthritis and other joint disorders. A disintegrin and metalloproteinase with thrombospondin motifs-5 (ADAMTS-5) is an aggrecanase that dominates the catalysis of aggrecan, the major proteoglycan in the cartilage ECM. Intriguingly, unlike its critical cleavage site Glu373-374Ala, another potential cleavage site, Glu419-420Ala, composed of the same amino acids in the aggrecan interglobular domain, is not a major cleavage site. It remains unclear how ADAMTS-5 distinguishes between them and hydrolyzes the correct scissile bonds. This research introduces a bottom-up in silico approach to reveal the molecular mechanism by which ADAMTS-5 recognizes the cleavage site on aggrecan. It is hypothesized that the sequence in the vicinity assists ADAMTS-5 in positioning the cleavage site. Specific residues were found to serve as binding sites, helping aggrecan bind more stably and fit into the enzyme better. The findings provide insight into the substrate binding and recognition mechanism for cartilage ECM degradation from a brand new atomic-scale perspective, laying the foundation for prophylaxis and treatment of related joint diseases.
Collapse
Affiliation(s)
- Yen-Yu Lai
- National Taiwan University, Department of Civil Engineering, Taipei 10617, Taiwan
| | - Deng Li
- National Taiwan University, Department of Civil Engineering, Taipei 10617, Taiwan
| | - Shu-Wei Chang
- National Taiwan University, Department of Civil Engineering, Taipei 10617, Taiwan
- National Taiwan University, Department of Biomedical Engineering, Taipei 10617, Taiwan
| |
Collapse
|
49
|
Muderspach SJ, Fredslund F, Volf V, Poulsen JCN, Blicher TH, Clausen MH, Rasmussen KK, Krogh KBRM, Jensen K, Lo Leggio L. Engineering the substrate binding site of the hyperthermostable archaeal endo-β-1,4-galactanase from Ignisphaera aggregans. BIOTECHNOLOGY FOR BIOFUELS 2021; 14:183. [PMID: 34530892 PMCID: PMC8447715 DOI: 10.1186/s13068-021-02025-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/23/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Endo-β-1,4-galactanases are glycoside hydrolases (GH) from the GH53 family belonging to the largest clan of GHs, clan GH-A. GHs are ubiquitous and involved in a myriad of biological functions as well as being widely used industrially. Endo-β-1,4-galactanases, in particular hydrolyse galactan and arabinogalactan in pectin, a major component of the primary plant cell wall, with important functions in plant defence and application in the food and other industries. Here, we explore the family's biological diversity by characterizing the first archaeal and hyperthermophilic GH53 galactanase, and utilize it as a scaffold for engineering enzymes with different product lengths. RESULTS A galactanase gene was identified in the genome of the anaerobic hyperthermophilic archaeon Ignisphaera aggregans, and the isolated catalytic domain expressed and characterized (IaGal). IaGal presents the typical (βα)8 barrel structure of clan GH-A enzymes, with catalytic carboxylates at the end of the 4th and 7th barrel strands. Its activity optimum of at least 95 °C and melting point over 100 °C indicate extreme thermostability, a very advantageous property for industrial applications. If enzyme depletion is reduced, so is the need for re-addition, and thus costs. The main stabilizing features of IaGal compared to other structurally characterized members are π-π and cation-π interactions. The length of the substrate binding site-and thus produced oligosaccharide products-is intermediate compared to previously characterized galactanases. Variants inspired by the structural diversity in the GH53 family were rationally designed to shorten or extend the substrate binding groove, in order to modulate product length. Subsite-deleted variants produced shorter products than IaGal, as do the fungal galactanases inspiring the design. IaGal variants engineered with a longer binding site produced a less expected degradation pattern, though still different from that of wild-type IaGal. All variants remained extremely stable. CONCLUSIONS We have characterized in detail the most thermophilic endo-β-1,4-galactanase known to date and successfully engineered it to modify the degradation profile, while maintaining much of its desirable thermostability. This is an important achievement as oligosaccharide products length is an important property for industrial and natural GHs alike.
Collapse
Affiliation(s)
- Sebastian J Muderspach
- Department of Chemistry, University of Copenhagen, Universitetsparken 5, 2100, Copenhagen, Denmark
| | - Folmer Fredslund
- Department of Chemistry, University of Copenhagen, Universitetsparken 5, 2100, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kgs. Lyngby, Denmark
| | - Verena Volf
- Novozymes A/S, Biologiens vej 2, 2800, Kongens Lyngby, Denmark
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | | | | | - Mads Hartvig Clausen
- Center for Nanomedicine and Theranostics, Department of Chemistry, Technical University of Denmark, Kemitorvet 207, 2800, Kgs. Lyngby, Denmark
| | - Kim Krighaar Rasmussen
- Department of Chemistry, University of Copenhagen, Universitetsparken 5, 2100, Copenhagen, Denmark
| | | | - Kenneth Jensen
- Novozymes A/S, Biologiens vej 2, 2800, Kongens Lyngby, Denmark.
| | - Leila Lo Leggio
- Department of Chemistry, University of Copenhagen, Universitetsparken 5, 2100, Copenhagen, Denmark.
| |
Collapse
|
50
|
Nanomolar inhibition of human OGA by 2-acetamido-2-deoxy-d-glucono-1,5-lactone semicarbazone derivatives. Eur J Med Chem 2021; 223:113649. [PMID: 34186233 DOI: 10.1016/j.ejmech.2021.113649] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/11/2021] [Accepted: 06/12/2021] [Indexed: 12/25/2022]
Abstract
O-GlcNAcylation is a dynamic post-translational modification mediated by O-linked β-N-acetylglucosamine transferase (OGT) and O-GlcNAc hydrolase (OGA), that adds or removes a single β-N-acetylglucosamine (GlcNAc) moiety to or from serine/threonine residues of nucleocytosolic and mitochondrial proteins, respectively. The perturbed homeostasis of O-GlcNAc cycling results in several pathological conditions. Human OGA is a promising therapeutic target in diseases where aberrantly low levels of O-GlcNAc are experienced, such as tauopathy in Alzheimer's disease. A new class of potent OGA inhibitors, 2-acetamido-2-deoxy-d-glucono-1,5-lactone (thio)semicarbazones, have been identified. Eight inhibitors were designed and synthesized in five steps starting from d-glucosamine and with 15-55% overall yields. A heterologous OGA expression protocol with strain selection and isolation has been optimized that resulted in stable, active and full length human OGA (hOGA) isomorph. Thermal denaturation kinetics of hOGA revealed environmental factors affecting hOGA stability. From kinetics experiments, the synthesized compounds proved to be efficient competitive inhibitors of hOGA with Ki-s in the range of ∼30-250 nM and moderate selectivity with respect to lysosomal β-hexosaminidases. In silico studies consisting of Prime protein-ligand refinements, QM/MM optimizations and QM/MM-PBSA binding free energy calculations revealed the factors governing the observed potencies, and led to design of the most potent analogue 2-acetamido-2-deoxy-d-glucono-1,5-lactone 4-(2-naphthyl)-semicarbazone 6g (Ki = 36 nM). The protocol employed has applications in future structure based inhibitor design targeting OGA.
Collapse
|