1
|
Elhanafy E, Akbari Ahangar A, Roth R, Gamal El-Din TM, Bankston JR, Li J. The differential impacts of equivalent gating-charge mutations in voltage-gated sodium channels. J Gen Physiol 2025; 157:e202413669. [PMID: 39820972 PMCID: PMC11740781 DOI: 10.1085/jgp.202413669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 11/27/2024] [Accepted: 12/25/2024] [Indexed: 01/19/2025] Open
Abstract
Voltage-gated sodium (Nav) channels are pivotal for cellular signaling, and mutations in Nav channels can lead to excitability disorders in cardiac, muscular, and neural tissues. A major cluster of pathological mutations localizes in the voltage-sensing domains (VSDs), resulting in either gain-of-function, loss-of-function effects, or both. However, the mechanism behind this functional diversity of mutations at equivalent positions remains elusive. Through hotspot analysis, we identified three gating charges (R1, R2, and R3) as major mutational hotspots in VSDs. The same amino acid substitutions at equivalent gating-charge positions in VSDI and VSDII of the cardiac sodium channel Nav1.5 show differential gating property impacts in electrophysiology measurements. We conducted molecular dynamics (MD) simulations on wild-type channels and six mutants to elucidate the structural basis of their differential impacts. Our 120-µs MD simulations with applied external electric fields captured VSD state transitions and revealed the differential structural dynamics between equivalent R-to-Q mutants. Notably, we observed transient leaky conformations in some mutants during structural transitions, offering a detailed structural explanation for gating-pore currents. Our salt-bridge network analysis uncovered VSD-specific and state-dependent interactions among gating charges, countercharges, and lipids. This detailed analysis revealed how mutations disrupt critical electrostatic interactions, thereby altering VSD permeability and modulating gating properties. By demonstrating the crucial importance of considering the specific structural context of each mutation, our study advances our understanding of structure-function relationships in Nav channels. Our work establishes a robust framework for future investigations into the molecular basis of ion channel-related disorders.
Collapse
Affiliation(s)
- Eslam Elhanafy
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS, USA
| | - Amin Akbari Ahangar
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS, USA
| | - Rebecca Roth
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - John R Bankston
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jing Li
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS, USA
| |
Collapse
|
2
|
Lagunas A, Belloir C, Lalis M, Briand L, Topin J, Gorostiza P, Samitier J. Ligand discrimination in hOR1A1 based on the capacitive response. Biosens Bioelectron 2025; 271:117000. [PMID: 39626347 DOI: 10.1016/j.bios.2024.117000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 01/06/2025]
Abstract
Odorant discrimination mechanisms are based on the differential interactions between odorant molecules and olfactory receptors (ORs). Biohybrid sensors based on ORs described to date show selectivity towards specific versus non-specific binding of odorants, being unable to distinguish between specific ligands of different affinity. Here we disclose a method that enables odorant discrimination based on the modulation of the capacitive response of the receptor, which allows the differentiation of three high-affinity hOR1A1 agonists. We performed voltammetry and impedance measurements of the hOR1A1 receptor selectively immobilized on a gold electrode in the absence and presence of the agonists. Binding induces a decrease in the capacitive response of the receptor that is proportional to the ligand potency, reaching up to a 40% decrease for the cognate ligand dihydrojasmone, which is attributed to changes in the magnitude and orientation of the electric dipole in the receptor, thereby regulating its response to the applied electric field.
Collapse
Affiliation(s)
- Anna Lagunas
- CIBER-BBN, ISCIII, Madrid, Spain; Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, C/Baldiri i Reixac 10-12, 08028, Barcelona, Spain.
| | - Christine Belloir
- Centre des Sciences Du Goût et de L'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, 9E Bd Jeanne D'Arc, F-21000, Dijon, France
| | - Maxence Lalis
- Institut de Chimie de Nice, Université Côte D'Azur, Campus Sciences, Parc Valrose 28 avenue, Valrose, 06108, Nice, Cedex 2, France
| | - Loïc Briand
- Centre des Sciences Du Goût et de L'Alimentation, CNRS, INRAE, Institut Agro, Université de Bourgogne, 9E Bd Jeanne D'Arc, F-21000, Dijon, France
| | - Jérémie Topin
- Institut de Chimie de Nice, Université Côte D'Azur, Campus Sciences, Parc Valrose 28 avenue, Valrose, 06108, Nice, Cedex 2, France
| | - Pau Gorostiza
- CIBER-BBN, ISCIII, Madrid, Spain; Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, C/Baldiri i Reixac 10-12, 08028, Barcelona, Spain; Catalan Institution for Research and Advanced Studies (ICREA), pg. Lluís Companys 23, 08010, Barcelona, Spain
| | - Josep Samitier
- CIBER-BBN, ISCIII, Madrid, Spain; Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology, C/Baldiri i Reixac 10-12, 08028, Barcelona, Spain; Department of Electronics and Biomedical Engineering, Faculty of Physics, University of Barcelona (UB), C/Martí i Franquès 1, 08028, Barcelona, Spain
| |
Collapse
|
3
|
Sun ZY, Liang T, Zhang Y, Hou G, Chu X, Hou JZ, Li W, Xie XQ, Feng Z. Structural insight into CD20/CD3-bispecific antibodies by molecular modeling. Comput Biol Med 2025; 185:109497. [PMID: 39674067 DOI: 10.1016/j.compbiomed.2024.109497] [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: 07/30/2024] [Revised: 11/09/2024] [Accepted: 11/26/2024] [Indexed: 12/16/2024]
Abstract
Non-Hodgkin's Lymphoma (NHL) remains a significant challenge in hematology, with chemotherapy and radiation therapy as conventional treatment options, albeit with limitations such as adverse effects. Immunotherapy, particularly bispecific antibodies (BsAbs) T cell engagers (TCEs), has emerged as a promising approach. Despite their potential, TCEs pose challenges, including adverse events like cytokine release syndrome. Understanding the structural details of TCEs and their interactions with target proteins is crucial for optimizing their therapeutic efficacy and toxicity. In this study, we further developed our protocol MCCS-Docker for protein-protein interactions and applied it to investigate the structural intricacies of CD3 interactions with therapeutic antibodies such as OKT3, UCHT1, Mosunetuzumab, Odronextumab, Glofitamab, and Epcoritamab using computational modeling techniques. Our analysis not only approved the effectiveness of our updated MCCS-Docker protocol but also revealed detailed binding interactions between the BsAbs and CD3, elucidating key residues of Tyrosine and Asparagine in the antibodies involved in the binding interface. Molecular dynamics simulations validated the stability of these interactions over time, confirming the reliability of the binding poses generated from docking studies. Overall, our study offered a novel method to predict critical residues in protein-protein interactions and enhanced the understanding of the structural determinants governing BsAb interactions with target proteins, offering valuable insights for designing and optimizing immunotherapeutic agents for NHL and related hematologic malignancies.
Collapse
Affiliation(s)
- Ze-Yu Sun
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, and Pharmacometrics & System Pharmacology PharmacoAnalytics, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| | - Tianjian Liang
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, and Pharmacometrics & System Pharmacology PharmacoAnalytics, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| | - Yiyang Zhang
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, and Pharmacometrics & System Pharmacology PharmacoAnalytics, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| | - GanQian Hou
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, and Pharmacometrics & System Pharmacology PharmacoAnalytics, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| | - Xiaojie Chu
- Department of Medicine, Center for Antibody Therapeutics, Division of Infectious Diseases, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.
| | - Jing-Zhou Hou
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA15232, United States.
| | - Wei Li
- Department of Medicine, Center for Antibody Therapeutics, Division of Infectious Diseases, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States.
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, and Pharmacometrics & System Pharmacology PharmacoAnalytics, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| | - Zhiwei Feng
- Department of Pharmaceutical Sciences, Computational Chemical Genomics Screening Center, and Pharmacometrics & System Pharmacology PharmacoAnalytics, School of Pharmacy, National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| |
Collapse
|
4
|
Finke F, Hungerland J, Solov'yov IA, Schuhmann F. Different receptor models show differences in ligand binding strength and location: a computational drug screening for the tick-borne encephalitis virus. Mol Divers 2025; 29:281-292. [PMID: 38739227 PMCID: PMC11785706 DOI: 10.1007/s11030-024-10850-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/16/2024] [Indexed: 05/14/2024]
Abstract
The tick-borne encephalitis virus (TBE) is a neurotrophic disease that has spread more rapidly throughout Europe and Asia in the past few years. At the same time, no cure or specific therapy is known to battle the illness apart from vaccination. To find a pharmacologically relevant drug, a computer-aided drug screening was initiated. Such a procedure probes a possible binding of a drug to the RNA Polymerase of TBE. The crystal structure of the receptor, however, includes missing and partially modeled regions, which rendered the structure incomplete and of questionable use for a thorough drug screening procedure. The quality of the receptor model was addressed by studying three putative structures created. We show that the choice of receptor models greatly influences the binding affinity of potential drug molecules and that the binding location could also be significantly impacted. We demonstrate that some drug candidates are unsuitable for one model but show decent results for another. Without any prejudice on the three employed receptor models, the study reveals the imperative need to investigate the receptor structure before drug binding is probed whether experimentally or computationally.
Collapse
Affiliation(s)
- Felicitas Finke
- Institute of Physics, Carl von Ossietzky Universität, Carl-von-Ossietzky-Str. 9-11, 26129, Oldenburg, Germany
| | - Jonathan Hungerland
- Institute of Physics, Carl von Ossietzky Universität, Carl-von-Ossietzky-Str. 9-11, 26129, Oldenburg, Germany
| | - Ilia A Solov'yov
- Institute of Physics, Carl von Ossietzky Universität, Carl-von-Ossietzky-Str. 9-11, 26129, Oldenburg, Germany.
- Research Centre for Neurosensory Science, Carl von Ossietzky Universität, Carl-von-Ossietzky-Str. 9-11, 26129, Oldenburg, Germany.
- Center for Nanoscale Dynamics (CENAD), Carl von Ossietzky Universität, Ammerländer Heerstr. 114-118, 26129, Oldenburg, Germany.
| | - Fabian Schuhmann
- Niels Bohr International Academy, Niels Bohr Institute, University of Copenhagen, Blegdamsvej 17, 2100, Copenhagen, Denmark.
| |
Collapse
|
5
|
Morales AE, Dong Y, Brown T, Baid K, Kontopoulos DG, Gonzalez V, Huang Z, Ahmed AW, Bhuinya A, Hilgers L, Winkler S, Hughes G, Li X, Lu P, Yang Y, Kirilenko BM, Devanna P, Lama TM, Nissan Y, Pippel M, Dávalos LM, Vernes SC, Puechmaille SJ, Rossiter SJ, Yovel Y, Prescott JB, Kurth A, Ray DA, Lim BK, Myers E, Teeling EC, Banerjee A, Irving AT, Hiller M. Bat genomes illuminate adaptations to viral tolerance and disease resistance. Nature 2025:10.1038/s41586-024-08471-0. [PMID: 39880942 DOI: 10.1038/s41586-024-08471-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 11/28/2024] [Indexed: 01/31/2025]
Abstract
Zoonoses are infectious diseases transmitted from animals to humans. Bats have been suggested to harbour more zoonotic viruses than any other mammalian order1. Infections in bats are largely asymptomatic2,3, indicating limited tissue-damaging inflammation and immunopathology. To investigate the genomic basis of disease resistance, the Bat1K project generated reference-quality genomes of ten bat species, including potential viral reservoirs. Here we describe a systematic analysis covering 115 mammalian genomes that revealed that signatures of selection in immune genes are more prevalent in bats than in other mammalian orders. We found an excess of immune gene adaptations in the ancestral chiropteran branch and in many descending bat lineages, highlighting viral entry and detection factors, and regulators of antiviral and inflammatory responses. ISG15, which is an antiviral gene contributing to hyperinflammation during COVID-19 (refs. 4,5), exhibits key residue changes in rhinolophid and hipposiderid bats. Cellular infection experiments show species-specific antiviral differences and an essential role of protein conjugation in antiviral function of bat ISG15, separate from its role in secretion and inflammation in humans. Furthermore, in contrast to humans, ISG15 in most rhinolophid and hipposiderid bats has strong anti-SARS-CoV-2 activity. Our work reveals molecular mechanisms that contribute to viral tolerance and disease resistance in bats.
Collapse
Affiliation(s)
- Ariadna E Morales
- LOEWE Centre for Translational Biodiversity Genomics, Frankfurt, Germany
- Senckenberg Research Institute, Frankfurt, Germany
- Faculty of Biosciences, Goethe-University, Frankfurt, Germany
| | - Yue Dong
- Deanery of Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Thomas Brown
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- DRESDEN concept Genome Center, Dresden, Germany
| | - Kaushal Baid
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Dimitrios -Georgios Kontopoulos
- LOEWE Centre for Translational Biodiversity Genomics, Frankfurt, Germany
- Senckenberg Research Institute, Frankfurt, Germany
- Faculty of Biosciences, Goethe-University, Frankfurt, Germany
| | - Victoria Gonzalez
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Zixia Huang
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland
| | - Alexis-Walid Ahmed
- LOEWE Centre for Translational Biodiversity Genomics, Frankfurt, Germany
- Senckenberg Research Institute, Frankfurt, Germany
- Faculty of Biosciences, Goethe-University, Frankfurt, Germany
| | - Arkadeb Bhuinya
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Leon Hilgers
- LOEWE Centre for Translational Biodiversity Genomics, Frankfurt, Germany
- Senckenberg Research Institute, Frankfurt, Germany
- Faculty of Biosciences, Goethe-University, Frankfurt, Germany
| | - Sylke Winkler
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- DRESDEN concept Genome Center, Dresden, Germany
| | - Graham Hughes
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland
| | - Xiaomeng Li
- Deanery of Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Ping Lu
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Yixin Yang
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Bogdan M Kirilenko
- LOEWE Centre for Translational Biodiversity Genomics, Frankfurt, Germany
- Senckenberg Research Institute, Frankfurt, Germany
- Faculty of Biosciences, Goethe-University, Frankfurt, Germany
| | - Paolo Devanna
- Neurogenetics of Vocal Communication Group, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
| | - Tanya M Lama
- Department of Ecology and Evolution, SUNY Stony Brook, Stony Brook, NY, USA
- Department of Biological Sciences, Smith College, Northampton, MA, USA
| | - Yomiran Nissan
- School of Zoology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Martin Pippel
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- DRESDEN concept Genome Center, Dresden, Germany
| | - Liliana M Dávalos
- Department of Ecology and Evolution, SUNY Stony Brook, Stony Brook, NY, USA
- Consortium for Inter-Disciplinary Environmental Research, SUNY Stony Brook, Stony Brook, NY, USA
| | - Sonja C Vernes
- Neurogenetics of Vocal Communication Group, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
- School of Biology, University of St Andrews, St Andrews, UK
| | - Sebastien J Puechmaille
- Institut Universitaire de France, Paris, France
- ISEM, University of Montpellier, CNRS, IRD, Montpellier, France
| | - Stephen J Rossiter
- School of Biological and Behavioural Sciences, Queen Mary University of London, London, UK
| | - Yossi Yovel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Consortium for Inter-Disciplinary Environmental Research, SUNY Stony Brook, Stony Brook, NY, USA
| | - Joseph B Prescott
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Andreas Kurth
- Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - David A Ray
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
| | - Burton K Lim
- Department of Natural History, Royal Ontario Museum, Toronto, Ontario, Canada
| | - Eugene Myers
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- DRESDEN concept Genome Center, Dresden, Germany
| | - Emma C Teeling
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland
| | - Arinjay Banerjee
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Aaron T Irving
- Department of Infectious Diseases, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Center for Infection, Immunity and Cancer, Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China.
- Department of Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK.
| | - Michael Hiller
- LOEWE Centre for Translational Biodiversity Genomics, Frankfurt, Germany.
- Senckenberg Research Institute, Frankfurt, Germany.
- Faculty of Biosciences, Goethe-University, Frankfurt, Germany.
| |
Collapse
|
6
|
Grajales-Hernández DA, Roca M, Moliner V, López-Gallego F. Active Site Engineering of a Glycerol Dehydrogenase Improves its Oxidative Activity and Scope Toward Glycerol Derivatives. Chemistry 2025; 31:e202403735. [PMID: 39665443 DOI: 10.1002/chem.202403735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/29/2024] [Accepted: 12/11/2024] [Indexed: 12/13/2024]
Abstract
Regioselective oxidation of glyceryl alkyl ethers is of utmost importance for the fabrication of substituted hydroxy ketones and enantiopure 1,2-diols as green solvents and pharmaceutical building blocks, respectively. An engineered glycerol dehydrogenase from Bacillus stearothermophilus was described to perform the regioselective oxidation of alkyl glycerol ethers, identifying position 252 as key for accepting larger substrates than glycerol. In this work, we further engineer that position through partial saturation mutagenesis to broaden the substrate scope toward other glycerol derivatives, improving enzyme kinetics and minimizing product inhibition. In particular, the BsGlyDH-L252S variant becomes the most efficient biocatalyst for the deracemization of alkyl glyceryl ethers in a two-step, one-pot immobilized system. The discovery and use of these alternative mutants of GlyDH opens the road to more applications and increases the enzymatic toolbox for the modifications of glyceryl ethers.
Collapse
Affiliation(s)
- Daniel A Grajales-Hernández
- Heterogeneous Biocatalysis Laboratory, CICbiomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramón, 194, Donostia-San Sebastián, 20014, Spain
| | - Maite Roca
- Institute of Advanced Materials (INAM), Universitat Jaume I, Av. Vicent Sos Baynat, s/n, Castelló de la Plana, 12071, Spain
| | - Vicent Moliner
- Institute of Advanced Materials (INAM), Universitat Jaume I, Av. Vicent Sos Baynat, s/n, Castelló de la Plana, 12071, Spain
| | - Fernando López-Gallego
- Heterogeneous Biocatalysis Laboratory, CICbiomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramón, 194, Donostia-San Sebastián, 20014, Spain
- IKERBASQUE, Basque Foundation for Science, Maria Diaz de Haro 3, Bilbao, 48013, Spain
| |
Collapse
|
7
|
Wei W, Li X, Hou N, Xie A, Liang H, Gao T, Jing X, Li L, Hao J, Xu X. Novel inhibitors of the (VIBVN) NAT protein identified through pharmacophore modeling. Sci Rep 2025; 15:2898. [PMID: 39843504 PMCID: PMC11754874 DOI: 10.1038/s41598-025-85869-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025] Open
Abstract
Arylamine N-acetyltransferases (NATs, E.C. 2.3.1.5) constitute a family of phase II drug metabolizing enzymes. These enzymes catalyze the transfer of acetyl groups from acetyl-CoA to a variety of substrates including arylamines, arylhydrazines, and N-hydroxyarylamines. By facilitating these reactions, NATs play a pivotal role in the detoxification and metabolic processing of a wide range of drugs and carcinogens. NAT in marine V. vulnificus plays a role in the metabolism of drugs, leading to the development of drug resistance in marine V. vulnificus. However, inhibitors targeted marine V. vulnificus NAT [(VIBVN)NAT] remain unclear. Therefore, our research aimed to identify potential hit compounds that target (VIBVN)NAT. We integrated multiple computational approaches to screen for effective inhibitors. From this process, we identified two hit compounds, AK-968-11563024 and AG-205-36710025, with IC50 values of 18.86 µM and 33.27 µM, respectively. Molecular dynamics simulations further elucidated the binding mechanism between (VIBVN)NAT and AK-968-11563024. Our study revealed that AK-968-11563024 forms stable interactions with PHE124, HIS167, and TRP230, which may contribute to its biological activity. Our findings provide a valuable foundation for the future development of drugs targeted therapeutics against (VIBVN)NAT.
Collapse
Affiliation(s)
- Wei Wei
- Affiliated Huzhou Hospital, The Key Laboratory of Molecular Medicine, Zhejiang University School of Medicine, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, 313000, China
- Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
| | - Xionghao Li
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ministry of Education, Ocean University of China, Qingdao, 266071, China
- Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China
- Network and Information Center, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Ning Hou
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ministry of Education, Ocean University of China, Qingdao, 266071, China
| | - Aowei Xie
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266071, China
| | - Huicong Liang
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ministry of Education, Ocean University of China, Qingdao, 266071, China
| | - Ting Gao
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266071, China
| | - Xiaoli Jing
- Network and Information Center, Qingdao Marine Science and Technology Center, Qingdao, 266237, China
| | - Liqin Li
- Affiliated Huzhou Hospital, The Key Laboratory of Molecular Medicine, Zhejiang University School of Medicine, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, 313000, China
- TCM Key Laboratory Cultivation Base of Zhe jiang Province for the Development and Clinical Transformation of Immunomodulatory drugs, Huzhou Central Hospital, Huzhou, 313000, China
| | - Jiejie Hao
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ministry of Education, Ocean University of China, Qingdao, 266071, China
| | - Ximing Xu
- Key Laboratory of Marine Drugs, School of Medicine and Pharmacy, Ministry of Education, Ocean University of China, Qingdao, 266071, China.
- Marine Biomedical Research Institute of Qingdao, Qingdao, 266071, China.
- Network and Information Center, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| |
Collapse
|
8
|
Min SM, Bashore FM, Smith JL, Havener TM, Howell S, Li H, Couñago RM, Popov KI, Axtman AD. Development of a Second-Generation, In Vivo Chemical Probe for PIKfyve. J Med Chem 2025. [PMID: 39838960 DOI: 10.1021/acs.jmedchem.4c02531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
We optimized our highly potent and cell-active chemical probe for phosphatidylinositol-3-phosphate 5-kinase (PIKfyve), SGC-PIKFYVE-1, resulting in compounds with improved potency and demonstrated in vivo stability. Use of an in-cell, kinome-wide selectivity panel allowed for confirmation of excellent in-cell selectivity of our lead compound, 40, and another promising analogue, 46. Evaluation of the pharmacokinetic (PK) profiles of these two compounds revealed that both are well tolerated systemically and orally bioavailable. Coupled with its subnanomolar cellular potency and impressive selectivity in cells, the long half-life of 40 makes it an ideal candidate for the evaluation of the consequences of PIKfyve inhibition in vivo. PIKfyve inhibition has been investigated clinically for indications including rheumatoid arthritis, Crohn's disease, COVID-19, and ALS using a single compound (apilimod), supporting the development of orthogonal PIKfyve inhibitors with in vivo stability.
Collapse
Affiliation(s)
- Sophia M Min
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Frances M Bashore
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Jeffery L Smith
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Tammy M Havener
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Stefanie Howell
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Haoxi Li
- Laboratory for Molecular Modeling, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rafael M Couñago
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, UNICAMP, Campinas, SP 13083-886, Brazil
| | - Konstantin I Popov
- Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Alison D Axtman
- Structural Genomics Consortium, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
9
|
Reinhardt CR, Lee JA, Hendricks L, Green T, Kunczynski L, Roberts AJ, Miller N, Rafalin N, Kulik HJ, Pollock CJ, Austin RN. No Bridge between Us: EXAFS and Computations Confirm Two Distant Iron Ions Comprise the Active Site of Alkane Monooxygenase (AlkB). J Am Chem Soc 2025; 147:2432-2443. [PMID: 39772501 PMCID: PMC11753938 DOI: 10.1021/jacs.4c12633] [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] [Indexed: 01/11/2025]
Abstract
Alkane monooxygenase (AlkB) is the dominant enzyme that catalyzes the oxidation of liquid alkanes in the environment. Two recent structural models derived from cryo-electron microscopy (cryo-EM) reveal an unusual active site: a histidine-rich center that binds two iron ions without a bridging ligand. To ensure that potential photoreduction and radiation damage are not responsible for the absence of a bridging ligand in the cryo-EM structures, spectroscopic methods are needed. We present the results of extended X-ray absorption fine structure (EXAFS) experiments collected under conditions where photodamage was avoided. Careful data analysis reveals an active site structure consistent with the cryo-EM structures in which the two iron ions are ligated by nine histidines and separated by at least 5 Å. The EXAFS data were used to inform structural models for molecular dynamics (MD) simulations. The MD simulations corroborate EXAFS observations that neither of the two conserved carboxylate-containing residues (E281 and D190) near the active site are likely candidates for metal ion bridging. Mutagenesis experiments, spectroscopy, and additional MD simulations were used to further explore the role of these carboxylate residues. A variant in which a carboxylate containing residue (E281) was changed to a methyl residue (E281A) showed little change in pre-edge features, consistent with the observation that it is not essential for activity and hence unlikely to serve as a bridging ligand at any point in the catalytic cycle. D190 variants had substantially diminished activity, suggesting an important role in catalysis not yet fully understood.
Collapse
Affiliation(s)
- Clorice R. Reinhardt
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Juliet A. Lee
- Department of Chemistry, Barnard College, 3009 Broadway, NY 10027 USA
| | - Lauren Hendricks
- Department of Chemistry, Barnard College, 3009 Broadway, NY 10027 USA
| | - Tierani Green
- Department of Chemistry, Barnard College, 3009 Broadway, NY 10027 USA
| | - Lily Kunczynski
- Department of Chemistry, Barnard College, 3009 Broadway, NY 10027 USA
| | | | - Naomi Miller
- Department of Chemistry, Barnard College, 3009 Broadway, NY 10027 USA
| | - Noga Rafalin
- Department of Chemistry, Barnard College, 3009 Broadway, NY 10027 USA
| | - Heather J. Kulik
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Christopher J. Pollock
- Cornell High Energy Synchrotron Source, Wilson Laboratory, Cornell University, Ithaca, NY 14853, USA
| | - Rachel N Austin
- Department of Chemistry, Barnard College, 3009 Broadway, NY 10027 USA
| |
Collapse
|
10
|
Nakazawa Y, Kageyama M, Matsuzawa T, Liang Z, Kobayashi K, Shimizu H, Maeda K, Masuhiro M, Motouchi S, Kumano S, Tanaka N, Kuramochi K, Nakai H, Taguchi H, Nakajima M. Structure and function of a β-1,2-galactosidase from Bacteroides xylanisolvens, an intestinal bacterium. Commun Biol 2025; 8:66. [PMID: 39820076 PMCID: PMC11739564 DOI: 10.1038/s42003-025-07494-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 01/08/2025] [Indexed: 01/19/2025] Open
Abstract
Galactosides are major carbohydrates that are found in plant cell walls and various prebiotic oligosaccharides. Studying the detailed biochemical functions of β-galactosidases in degrading these carbohydrates is important. In particular, identifying β-galactosidases with new substrate specificities could help in the production of potentially beneficial oligosaccharides. In this study, we identify a β-galactosidase with novel substrate specificity from Bacteroides xylanisolvens, an intestinal bacterium. The enzyme do not show hydrolytic activity toward natural β-galactosides during the first screening. However, when α-D-galactosyl fluoride (α-GalF) as a donor substrate and galactose or D-fucose as an acceptor substrate are incubated with a nucleophile mutant, reaction products are detected. The galactobiose produced from the α-GalF and galactose is identified as β-1,2-galactobiose using NMR. Kinetic analysis reveals that this enzyme effectively hydrolyzes β-1,2-galactobiose and β-1,2-galactotriose. In the complex structure with methyl β-galactopyranose as a ligand, the ligand is only located at subsite +1. The 2-hydroxy group and the anomeric methyl group of methyl β-galactopyranose faces in the direction of subsite -1 and the solvent, respectively. This observation is consistent with the substrate specificity of the enzyme regarding linkage position and chain length. Overall, we conclude that the enzyme is a β-galactosidase acting on β-1,2-galactooligosaccharides.
Collapse
Affiliation(s)
- Yutaka Nakazawa
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Masumi Kageyama
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Tomohiko Matsuzawa
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, 2393 Ikenobe, Miki, Kagawa, 761-0795, Japan
| | - Ziqin Liang
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Kaito Kobayashi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
- Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology (AIST), 2-4-7 Aomi, Tokyo, Koto-ku, 135-0064, Japan
| | - Hisaka Shimizu
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Kazuki Maeda
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Miho Masuhiro
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Sei Motouchi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Saika Kumano
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Nobukiyo Tanaka
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Kouji Kuramochi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Hiroyuki Nakai
- Faculty of Agriculture, Niigata University, 8050 Ikarashi 2-no-cho, Niigata, Nishi-ku, 950-2181, Japan
| | - Hayao Taguchi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan
| | - Masahiro Nakajima
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba, 278-8510, Japan.
| |
Collapse
|
11
|
Farag M, Guedeney N, Schwalen F, Zadoroznyj A, Barczyk A, Giret M, Antraygues K, Wang A, Cornu M, Suzanne P, Since M, Sophie Voisin-Chiret A, Dubrez L, Leleu-Chavain N, Kieffer C, Sopkova-de Oliveira Santos J. Towards New Anti-Inflammatory Agents: Design, Synthesis and Evaluation of Molecules Targeting XIAP-BIR2. ChemMedChem 2025; 20:e202400567. [PMID: 39364702 DOI: 10.1002/cmdc.202400567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/19/2024] [Accepted: 10/02/2024] [Indexed: 10/05/2024]
Abstract
The X-chromosome-linked inhibitor of apoptosis protein (XIAP) plays a crucial role in controlling cell survival across multiple regulated cell death pathways and coordinating a range of inflammatory signalling events. The discovery of selective inhibitors for XIAP-BIR2, able to disrupt the direct physical interaction between XIAP and RIPK2, offer promising therapeutic options for NOD2-mediated diseases like Crohn's disease, sarcoidosis, and Blau syndrome. The objective of this study was to design, synthesize, and evaluate small synthetic molecules with binding selectivity to XIAP-BIR2 domain. To achieve this, we applied an interdisciplinary drug design approach and firstly we have synthesized an initial fragment library to achieve a first XIAP inhibition activity. Then using a growing strategy, larger compounds were synthesized and one of them presents a good selectivity for XIAP-BIR2 versus XIAP-BIR3 domain, compound 20 c. The ability of compound 20 c to block the NOD1/2 pathway was confirmed in cell models. These data show that we have synthesized molecules capable of blocking NOD1/2 signalling pathways in cellulo, and ultimately leading to new anti-inflammatory compounds.
Collapse
Affiliation(s)
- Marc Farag
- Department, Normandie Univ, UNICAEN, CERMN, bd Becquerel, F-14000, Caen, Cedex, France
| | - Nicolas Guedeney
- Department, Normandie Univ, UNICAEN, CERMN, bd Becquerel, F-14000, Caen, Cedex, France
| | - Florian Schwalen
- Department, Normandie Univ, UNICAEN, CERMN, bd Becquerel, F-14000, Caen, Cedex, France
| | - Aymeric Zadoroznyj
- Institut National de la Santé et de la Recherche Médicale (Inserm), LNC UMR1231, Dijon, France
- Université de Bourgogne Franche-Comté, LNC UMR1231, Dijon, France
| | - Amélie Barczyk
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
| | - Martin Giret
- Department, Normandie Univ, UNICAEN, CERMN, bd Becquerel, F-14000, Caen, Cedex, France
| | - Kevin Antraygues
- Department, Normandie Univ, UNICAEN, CERMN, bd Becquerel, F-14000, Caen, Cedex, France
| | - Alice Wang
- Department, Normandie Univ, UNICAEN, CERMN, bd Becquerel, F-14000, Caen, Cedex, France
| | - Marie Cornu
- Department, Normandie Univ, UNICAEN, CERMN, bd Becquerel, F-14000, Caen, Cedex, France
| | - Peggy Suzanne
- Department, Normandie Univ, UNICAEN, CERMN, bd Becquerel, F-14000, Caen, Cedex, France
| | - Marc Since
- Department, Normandie Univ, UNICAEN, CERMN, bd Becquerel, F-14000, Caen, Cedex, France
| | | | - Laurence Dubrez
- Institut National de la Santé et de la Recherche Médicale (Inserm), LNC UMR1231, Dijon, France
- Université de Bourgogne Franche-Comté, LNC UMR1231, Dijon, France
| | - Natascha Leleu-Chavain
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, F-59000, Lille, France
| | - Charline Kieffer
- Department, Normandie Univ, UNICAEN, CERMN, bd Becquerel, F-14000, Caen, Cedex, France
| | | |
Collapse
|
12
|
Yan X, Zhu L, Li Q, Tian Y, Qiu J, Liu X, Tong HHY, Ouyang Q, Yao X, Liu H. QM/MM study reveals novel mechanism of KRAS and KRAS G12R catalyzed GTP hydrolysis. Int J Biol Macromol 2025; 297:139820. [PMID: 39805439 DOI: 10.1016/j.ijbiomac.2025.139820] [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: 12/04/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
As a crucial drug target, KRAS can regulate most cellular processes involving guanosine triphosphate (GTP) hydrolysis. However, the mechanism of GTP hydrolysis has remained controversial over the past decades. Here, several different GTP hydrolysis mechanisms catalyzed by wild-type KRAS (WT-KRAS) and KRASG12R mutants were discussed via four QM/MM calculation models. Based on the computational results, a Mg2+-coordinated H2O-mediated GTP hydrolysis mechanism was proposed. In this mechanism, a Mg2+-coordinated H2O first protonates the fully deprotonated GTP, and then the Mg2+ coordinated hydroxyl anion is generated. The Pγ-O bond is formed via the SN2 attack of the second H2O on the Pγ atom of the GTP, leading to the simultaneous cleavage of the Pγ-O bond. Meanwhile, the hydroxyl anion coordinated to Mg2+ and generated in the first step acts as a proton acceptor from water. This Mg2+ coordinated H2O-involved GTP hydrolysis mechanism may also be suitable for Mg2+-catalyzed ATP hydrolysis. Furthermore, the mechanism of GTP hydrolysis catalyzed by the KRASG12R mutant, whose hydrolysis rate was approximately 40-fold slower than WT-KRAS, was also discussed. Our QM/MM calculations reveal that GTP is easily protonated by the residue R12, and the energy barrier of GTP hydrolysis catalyzed by the KRASG12R mutant is lower than the corresponding barrier for WT-KRAS. Nevertheless, molecular dynamics (MD) simulations reveal that R12, a residue characterized by significant steric hindrance, is positioned at the GTP site where the nucleophilic attack by water occurs during Pγ-O bond formation, thereby strongly impeding the approach of water molecules to GTP. As a result, the GTP hydrolysis rate catalyzed by the KRASG12R mutant was severely impaired. Uncovering the GTP hydrolysis mechanism catalyzed by the WT-KRAS and KRASG12R mutant may also give a reasonable explanation for the relationship between the KRASG12R mutation and the occurrence of cancer. We hope this finding will provide useful guidance for drug discovery that targets KRAS.
Collapse
Affiliation(s)
- Xiao Yan
- Faculty of Applied Sciences, Macao Polytechnic University, Macao, SAR, China
| | - Lei Zhu
- College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China
| | - Qin Li
- Faculty of Applied Sciences, Macao Polytechnic University, Macao, SAR, China
| | - Yanan Tian
- Faculty of Applied Sciences, Macao Polytechnic University, Macao, SAR, China
| | - Jiayue Qiu
- Faculty of Applied Sciences, Macao Polytechnic University, Macao, SAR, China
| | - Xiaomeng Liu
- Faculty of Applied Sciences, Macao Polytechnic University, Macao, SAR, China
| | - Henry H Y Tong
- Faculty of Applied Sciences, Macao Polytechnic University, Macao, SAR, China
| | - Qin Ouyang
- College of Pharmacy, Third Military Medical University, Shapingba, Chongqing 400038, China.
| | - Xiaojun Yao
- Faculty of Applied Sciences, Macao Polytechnic University, Macao, SAR, China.
| | - Huanxiang Liu
- Faculty of Applied Sciences, Macao Polytechnic University, Macao, SAR, China.
| |
Collapse
|
13
|
Balduzzi E, Yin W, Lambry JC, Myllykallio H, Aleksandrov A. Additive CHARMM Force Field for Pterins and Folates. J Comput Chem 2025; 46:e27548. [PMID: 39710837 DOI: 10.1002/jcc.27548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/06/2024] [Accepted: 11/15/2024] [Indexed: 12/24/2024]
Abstract
Folates comprise a crucial class of biologically active compounds related to folic acid, playing a vital role in numerous enzymatic reactions. One-carbon metabolism, facilitated by the folate cofactor, supports numerous physiological processes, including biosynthesis, amino acid homeostasis, epigenetic maintenance, and redox defense. Folates share a common pterin heterocyclic ring structure capable of undergoing redox reactions and existing in various protonation states. This study aimed to derive molecular mechanics (MM) parameters compatible with the CHARMM36 all-atom additive force field for pterins and biologically important folates, including pterin, biopterin, and folic acid. Three redox forms were considered: oxidized, dihydrofolate, and tetrahydrofolate states. Across all protonation states, a total of 18 folates were parameterized. Partial charges were derived using the CHARMM force field parametrization protocol, based on targeting reference quantum mechanics monohydrate interactions, electrostatic potential, and dipole moment. Bonded terms were parameterized using one-dimensional adiabatic potential energy surface scans, and two-dimensional scans to parametrize in-ring torsions associated with the puckering states of dihydropterin and tetrahydropterin. The quality of the model was demonstrated through simulations of three protein complexes using optimized and initial parameters. These simulations underscored the significantly enhanced performance of the folate model developed in this study compared to the initial model without optimization in reproducing structural properties of folate-protein complexes. Overall, the presented MM model will be valuable for modeling folates in various redox states and serve as a starting point for parameterizing other folate derivatives.
Collapse
Affiliation(s)
- Elsa Balduzzi
- Laboratoire d'Optique et Biosciences (CNRS UMR7645, INSERM U1182), Ecole Polytechnique, Institut polytechnique de Paris, Palaiseau, France
| | - Wenlu Yin
- Laboratoire d'Optique et Biosciences (CNRS UMR7645, INSERM U1182), Ecole Polytechnique, Institut polytechnique de Paris, Palaiseau, France
| | - Jean-Christophe Lambry
- Laboratoire d'Optique et Biosciences (CNRS UMR7645, INSERM U1182), Ecole Polytechnique, Institut polytechnique de Paris, Palaiseau, France
| | - Hannu Myllykallio
- Laboratoire d'Optique et Biosciences (CNRS UMR7645, INSERM U1182), Ecole Polytechnique, Institut polytechnique de Paris, Palaiseau, France
| | - Alexey Aleksandrov
- Laboratoire d'Optique et Biosciences (CNRS UMR7645, INSERM U1182), Ecole Polytechnique, Institut polytechnique de Paris, Palaiseau, France
| |
Collapse
|
14
|
Goyzueta-Mamani LD, Pagliara Lage D, Barazorda-Ccahuana HL, Paco-Chipana M, Candia-Puma MA, Davila-Del-Carpio G, Galdino AS, Machado-de-Avila RA, Cordeiro Giunchetti R, D’Antonio EL, Ferraz Coelho EA, Chávez-Fumagalli MA. Exploring the Potential of Malvidin and Echiodinin as Probable Antileishmanial Agents Through In Silico Analysis and In Vitro Efficacy. Molecules 2025; 30:173. [PMID: 39795229 PMCID: PMC11722285 DOI: 10.3390/molecules30010173] [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: 12/02/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Leishmaniasis, a neglected tropical disease caused by Leishmania species, presents serious public health challenges due to limited treatment options, toxicity, high costs, and drug resistance. In this study, the in vitro potential of malvidin and echioidinin is examined as antileishmanial agents against L. amazonensis, L. braziliensis, and L. infantum, comparing their effects to amphotericin B (AmpB), a standard drug. Malvidin demonstrated greater potency than echioidinin across all parasite stages and species. Against L. amazonensis, malvidin's IC50 values were 197.71 ± 17.20 µM (stationary amastigotes) and 258.07 ± 17 µM (axenic amastigotes), compared to echioidinin's 272.99 ± 29.90 μM and 335.96 ± 19.35 μM. AmpB was more potent, with IC50 values of 0.06 ± 0.01 µM and 0.10 ± 0.03 µM. Malvidin exhibited lower cytotoxicity (CC50: 2920.31 ± 80.29 µM) than AmpB (1.06 ± 0.12 µM) and a favorable selectivity index. It reduced infection rates by 35.75% in L. amazonensis-infected macrophages. The in silico analysis revealed strong binding between malvidin and Leishmania arginase, with the residues HIS139 and PRO258 playing key roles. Gene expression analysis indicated malvidin's modulation of oxidative stress and DNA repair pathways, involving genes like GLO1 and APEX1. These findings suggest malvidin's potential as a safe, natural antileishmanial compound, warranting further in vivo studies to confirm its therapeutic efficacy and pharmacokinetics in animal models.
Collapse
Affiliation(s)
- Luis Daniel Goyzueta-Mamani
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru; (L.D.G.-M.); (H.L.B.-C.); (M.P.-C.); (M.A.C.-P.)
| | - Daniela Pagliara Lage
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (D.P.L.); (E.A.F.C.)
| | - Haruna Luz Barazorda-Ccahuana
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru; (L.D.G.-M.); (H.L.B.-C.); (M.P.-C.); (M.A.C.-P.)
| | - Margot Paco-Chipana
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru; (L.D.G.-M.); (H.L.B.-C.); (M.P.-C.); (M.A.C.-P.)
| | - Mayron Antonio Candia-Puma
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru; (L.D.G.-M.); (H.L.B.-C.); (M.P.-C.); (M.A.C.-P.)
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Arequipa 04000, Peru;
| | - Gonzalo Davila-Del-Carpio
- Facultad de Ciencias Farmacéuticas, Bioquímicas y Biotecnológicas, Universidad Católica de Santa María, Arequipa 04000, Peru;
| | - Alexsandro Sobreira Galdino
- Laboratório de Biotecnologia de Microrganismos, Universidade Federal São João Del-Rei, Divinópolis 35501-296, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Biotecnologia Industrial (INCT-BI), Distrito Federal, Brasilia 70070-010, Brazil
| | | | - Rodolfo Cordeiro Giunchetti
- Laboratório de Biologia das Interações Celulares, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil;
- Instituto Nacional de Ciência e Tecnologia de Doenças Tropicais (INCT-DT), Salvador 40110-160, Brazil
| | - Edward L. D’Antonio
- Department of Natural Sciences, University of South Carolina Beaufort, 1 University Boulevard, Bluffton, SC 29909, USA;
| | - Eduardo Antonio Ferraz Coelho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Brazil; (D.P.L.); (E.A.F.C.)
| | - Miguel Angel Chávez-Fumagalli
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Católica de Santa María, Arequipa 04000, Peru; (L.D.G.-M.); (H.L.B.-C.); (M.P.-C.); (M.A.C.-P.)
| |
Collapse
|
15
|
Whitehead JN, Leferink NGH, Hay S, Scrutton NS. Determinants of Product Outcome in Two Sesquiterpene Synthases from the Thermotolerant Bacterium Rubrobacter radiotolerans. Chembiochem 2025; 26:e202400672. [PMID: 39400489 DOI: 10.1002/cbic.202400672] [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: 08/14/2024] [Revised: 10/09/2024] [Accepted: 10/14/2024] [Indexed: 10/15/2024]
Abstract
Rubrobacter radiotolerans nerolidol synthase (NerS) and trans-α-bergamotene synthase (BerS) are among the first terpene synthases (TPSs) discovered from thermotolerant bacteria, and, despite sharing the same substrate, make terpenoid products with different carbon scaffolds. Here, the potential thermostability of NerS and BerS was investigated, and NerS was found to retain activity up to 55 °C. A library of 22 NerS and BerS variants was designed to probe the differing reaction mechanisms of NerS and BerS, including residues putatively involved in substrate sequestration, cation-π stabilisation of reactive intermediates, and shaping of the active site contour. Two BerS variants showed improved in vivo titres vs the WT enzyme, and also yielded different ratios of the related sesquiterpenoids (E)-β-farnesene and trans-α-bergamotene. BerS-L86F was proposed to encourage substrate isomerisation by cation-π stabilisation of the first cationic intermediate, resulting in a greater proportion of trans-α-bergamotene. By contrast, BerS-S82L significantly preferred (E)-β-farnesene formation, attributed to steric blocking of the isomerisation step, consistent with what has been observed in several plant TPSs. Our work highlights the importance of isomerisation as a key determinant of product outcome in TPSs, and shows how a combined computational and experimental approach can characterise TPSs and variants with improved and altered functionality.
Collapse
Affiliation(s)
- Joshua N Whitehead
- Future, Biomanufacturing Research Hub, Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, University of Manchester, 131 Princess Street, Manchester, M7 7DN, UK
| | - Nicole G H Leferink
- Future, Biomanufacturing Research Hub, Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, University of Manchester, 131 Princess Street, Manchester, M7 7DN, UK
| | - Sam Hay
- Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, University of Manchester, 131 Princess Street, Manchester, M7 7DN, UK
| | - Nigel S Scrutton
- Future, Biomanufacturing Research Hub, Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, University of Manchester, 131 Princess Street, Manchester, M7 7DN, UK
- Manchester Institute of Biotechnology, Department of Chemistry, School of Natural Sciences, University of Manchester, 131 Princess Street, Manchester, M7 7DN, UK
| |
Collapse
|
16
|
Li P, Shi M, Wang Y, Liu Q, Du X, Wang X. pH-Dependent Assembly and Stability of Toll-Like Receptor 3/dsRNA Signaling Complex: Insights from Constant pH Molecular Dynamics and Metadynamics Simulations. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411445. [PMID: 39520076 PMCID: PMC11714240 DOI: 10.1002/advs.202411445] [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: 09/17/2024] [Revised: 10/26/2024] [Indexed: 11/16/2024]
Abstract
The pH-dependent assembly of Toll-like receptors (TLRs), which triggers a threshold-like response, is a key principle in immune signaling. While crystallography has revealed the intricate structure of these assembly complexes, the mechanisms underlying their pH dependency remain unclear. Herein, constant pH simulations and metadynamics are employed to investigate the pH-dependent assembly and stability of the TLR3/dsRNA signaling complex. The findings demonstrate that system pH regulates complex assembly and stability by modulating the protonation and charge states of histidines. Histidines in TLR3 act as pH-dependent, positively charged binding sites that capture negatively charged dsRNA. Additionally, these histidines form a [H682⁺]-[E626⁻] dipole, facilitating the assembly of two TLR3 molecules into an antisymmetric dimer through dipole-dipole interactions. Surprisingly, TLR3 can shift the pKa values of key histidines from their model pKa of 6.5, increasing protonation likelihood and enhancing ligand binding. Notably, the aromatic residue Phe84, located within the dsRNA binding site [His39⁺-His60⁺-Phe84-His108⁺], alters the pKa of His60 through cation-π interactions with its protonated state. This study offers new insights into the molecular mechanisms underlying pH-dependent immune signaling via higher-order assemblies and suggests potential applications for histidine in self-assembling biomaterials.
Collapse
Affiliation(s)
- Penghui Li
- Shenzhen Key Laboratory of Marine Biotechnology and EcologyCollege of Life Sciences & OceanographyShenzhen UniversityShenzhen518055China
- Key Laboratory of Optoelectronic Devices and System of Ministry of Education and Guangdong ProvinceCollege Physics and Optoelectronic EngineeringShenzhen UniversityShenzhen518060China
| | - Mingsong Shi
- NHC Key Laboratory of Nuclear Technology Medical TransformationMianyang Central HospitalSchool of MedicineUniversity of Electronic Science and Technology of ChinaMianyangSichuan621099China
| | - Yibo Wang
- Laboratory of Chemical BiologyChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilin130022China
| | - Qiong Liu
- Shenzhen Key Laboratory of Marine Biotechnology and EcologyCollege of Life Sciences & OceanographyShenzhen UniversityShenzhen518055China
- Key Laboratory of Optoelectronic Devices and System of Ministry of Education and Guangdong ProvinceCollege Physics and Optoelectronic EngineeringShenzhen UniversityShenzhen518060China
- Shenzhen‐Hong Kong Institute of Brain ScienceShenzhen Fundamental Research InstitutionsShenzhen518055China
| | - Xiubo Du
- Shenzhen Key Laboratory of Marine Biotechnology and EcologyCollege of Life Sciences & OceanographyShenzhen UniversityShenzhen518055China
| | - Xiaohui Wang
- Laboratory of Chemical BiologyChangchun Institute of Applied ChemistryChinese Academy of SciencesChangchunJilin130022China
- School of Applied Chemistry and EngineeringUniversity of Science and Technology of ChinaHefei230026China
| |
Collapse
|
17
|
McIlroy PR, Pham LTM, Sheffield T, Stefan MA, Thatcher CE, Jaryenneh J, Schwedler JL, Sinha A, Sumner CA, Jones IKA, Won S, Bruneau RC, Weilhammer DR, Liu Z, Whelan S, Negrete OA, Sale KL, Harmon B. Nanobody screening and machine learning guided identification of cross-variant anti-SARS-CoV-2 neutralizing heavy-chain only antibodies. PLoS Pathog 2025; 21:e1012903. [PMID: 39847604 DOI: 10.1371/journal.ppat.1012903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 02/04/2025] [Accepted: 01/13/2025] [Indexed: 01/25/2025] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) continues to persist, demonstrating the risks posed by emerging infectious diseases to national security, public health, and the economy. Development of new vaccines and antibodies for emerging viral threats requires substantial resources and time, and traditional development platforms for vaccines and antibodies are often too slow to combat continuously evolving immunological escape variants, reducing their efficacy over time. Previously, we designed a next-generation synthetic humanized nanobody (Nb) phage display library and demonstrated that this library could be used to rapidly identify highly specific and potent neutralizing heavy chain-only antibodies (HCAbs) with prophylactic and therapeutic efficacy in vivo against the original SARS-CoV-2. In this study, we used a combination of high throughput screening and machine learning (ML) models to identify HCAbs with potent efficacy against SARS-CoV-2 viral variants of interest (VOIs) and concern (VOCs). To start, we screened our highly diverse Nb phage display library against several pre-Omicron VOI and VOC receptor binding domains (RBDs) to identify panels of cross-reactive HCAbs. Using HCAb affinity for SARS-CoV-2 VOI and VOCs (pre-Omicron variants) and model features from other published data, we were able to develop a ML model that successfully identified HCAbs with efficacy against Omicron variants, independent of our experimental biopanning workflow. This biopanning informed ML approach reduced the experimental screening burden by 78% to 90% for the Omicron BA.5 and Omicron BA.1 variants, respectively. The combined approach can be applied to other emerging viruses with pandemic potential to rapidly identify effective therapeutic antibodies against emerging variants.
Collapse
Affiliation(s)
- Peter R McIlroy
- Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, California, United States of America
| | - Le Thanh Mai Pham
- Bioresource and Environmental Security, Sandia National Laboratories, Livermore, California, United States of America
| | - Thomas Sheffield
- Biosecurity and Bioassurance, Sandia National Laboratories, Livermore, California, United States of America
| | - Maxwell A Stefan
- Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, California, United States of America
| | - Christine E Thatcher
- Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, California, United States of America
| | - James Jaryenneh
- Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, California, United States of America
| | - Jennifer L Schwedler
- Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, California, United States of America
| | - Anupama Sinha
- Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, California, United States of America
| | - Christopher A Sumner
- Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, California, United States of America
| | - Iris K A Jones
- Systems Biology, Sandia National Laboratories, Livermore, California, United States of America
| | - Stephen Won
- Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, California, United States of America
| | - Ryan C Bruneau
- Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, California, United States of America
| | - Dina R Weilhammer
- Biosciences and Biotechnology Division, Lawrence Livermore National Laboratories, Livermore, California, United States of America
| | - Zhuoming Liu
- Department of Molecular Microbiology, School of Medicine, Washington University, St. Louis, M issouri, United States of America
| | - Sean Whelan
- Department of Molecular Microbiology, School of Medicine, Washington University, St. Louis, M issouri, United States of America
| | - Oscar A Negrete
- Systems Biology, Sandia National Laboratories, Livermore, California, United States of America
| | - Kenneth L Sale
- Biosecurity and Bioassurance, Sandia National Laboratories, Livermore, California, United States of America
| | - Brooke Harmon
- Biotechnology and Bioengineering, Sandia National Laboratories, Livermore, California, United States of America
| |
Collapse
|
18
|
Haas-Neill L, Meneksedag-Erol D, Chaudhry A, Novoselova M, Ashraf QF, de Araujo ED, Wilson DJ, Rauscher S. The structural influence of the oncogenic driver mutation N642H in the STAT5B SH2 domain. Protein Sci 2025; 34:e70022. [PMID: 39723827 DOI: 10.1002/pro.70022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 12/28/2024]
Abstract
The point mutation N642H of the signal transducer and activator of transcription 5B (STAT5B) protein is associated with aggressive and drug-resistant forms of leukemia. This mutation is thought to promote cancer due to hyperactivation of STAT5B caused by increased stability of the active, parallel dimer state. However, the molecular mechanism leading to this stabilization is not well understood as there is currently no structure of the parallel dimer. To investigate the mutation's mechanism of action, we conducted extensive all-atom molecular dynamics simulations of multiple oligomeric forms of both STAT5B and STAT5BN642H, including a model for the parallel dimer. The N642H mutation directly affects the hydrogen bonding network within the phosphotyrosine (pY)-binding pocket of the parallel dimer, enhancing the pY-binding interaction. The simulations indicate that apo STAT5B is highly flexible, exploring a diverse conformational space. In contrast, apo STAT5BN642H accesses two distinct conformational states, one of which resembles the conformation of the parallel dimer. The simulation predictions of the effects of the mutation on structure and dynamics are supported by the results of hydrogen-deuterium exchange (HDX) mass spectrometry measurements carried out on STAT5B and STAT5BN642H in which a phosphopeptide was used to mimic the effects of parallel dimerization on the SH2 domain. The molecular-level information uncovered in this work contributes to our understanding of STAT5B hyperactivation by the N642H mutation and could help pave the way for novel therapeutic strategies targeting this mutation.
Collapse
Affiliation(s)
- Liam Haas-Neill
- Department of Physics, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Deniz Meneksedag-Erol
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Ayesha Chaudhry
- Department of Chemistry, York University, Toronto, Ontario, Canada
| | - Masha Novoselova
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Qirat F Ashraf
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Elvin D de Araujo
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada
| | - Derek J Wilson
- Department of Chemistry, York University, Toronto, Ontario, Canada
| | - Sarah Rauscher
- Department of Physics, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical and Physical Sciences, University of Toronto Mississauga, Mississauga, Ontario, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
19
|
Khan MW, Murali A. Modeling and interaction study of alcohol oxidase and ProteaseA in methylotrophic yeast C. boidinii: insights from In-silico analysis. J Biomol Struct Dyn 2024:1-17. [PMID: 39732624 DOI: 10.1080/07391102.2024.2446679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 10/21/2024] [Indexed: 12/30/2024]
Abstract
Flavin adenine nucleotide (FAD)-dependent oxidoreductase enzyme Alcohol oxidase (AOX) facilitates the growth of methylotrophic yeast C. boidinii by catabolizing methanol, producing formaldehyde and hydrogen peroxide. Vacuolar Protease-A (PrA) from C. boidinii is responsible for the proteolytic activity of AOX. However, no experimental structures for these enzymes have been reported. This in-silico study involves modeling and interaction analysis of AOX and PrA. A protein-protein interaction study shows that Thr75, Gly74, Arg72, Tyr73, and Met289 amino acids of PrA have shown interaction with AOX. These residues may be crucial for AOX proteolysis. An in-silico study predicts that serine protease inhibitors bind to specific amino acids, potentially obstructing PrA's degradable activity on AOX. PrA does not interact with the FAD binding sites in AOX. Instead, it interacts with AOX at sites (Ser337, Ala34, and Tyr343) where AOX monomers interact, hindering octamer formation the active form of AOX. During simulation, strong dynamics in PrA were found in the loop regions of the structure, as observed in the complexes. This in-silico work aims to corroborate the experimental research, which lacks structural studies on the proteolysis process.
Collapse
Affiliation(s)
- Mohammad Wahab Khan
- Department of Bioinformatics, School of Life Sciences Pondicherry University, Puducherry, India
| | - Ayaluru Murali
- Department of Bioinformatics, School of Life Sciences Pondicherry University, Puducherry, India
| |
Collapse
|
20
|
Rubini-Dias L, Fernandes TVA, de Souza MP, Hottz D, Arruda AT, Borges ADA, Ouverney G, da Silva FDC, Forezi LDSM, Limaverde-Sousa G, Robbs BK. Mannich Base Derived from Lawsone Inhibits PKM2 and Induces Neoplastic Cell Death. Biomedicines 2024; 12:2916. [PMID: 39767822 PMCID: PMC11673335 DOI: 10.3390/biomedicines12122916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: Pyruvate kinase M2, a central regulator of cancer cell metabolism, has garnered significant attention as a promising target for disrupting the metabolic adaptability of tumor cells. This study explores the potential of the Mannich base derived from lawsone (MB-6a) to interfere with PKM2 enzymatic activity both in vitro and in silico. Methods: The antiproliferative potential of MB-6a was tested using MTT assay in various cell lines, including SCC-9, Hep-G2, HT-29, B16-F10, and normal human gingival fibroblast (HGF). The inhibition of PKM2 mediated by MB-6a was assessed using an LDH-coupled assay and by measuring ATP production. Docking studies and molecular dynamics calculations were performed using Autodock 4 and GROMACS, respectively, on the tetrameric PKM2 crystallographic structure. Results: The Mannich base 6a demonstrated selective cytotoxicity against all cancer cell lines tested without affecting cell migration, with the highest selectivity index (SI) of 4.63 in SCC-9, followed by B16-F10 (SI = 3.9), Hep-G2 (SI = 3.4), and HT-29 (SI = 2.03). The compound effectively inhibited PKM2 glycolytic activity, leading to a reduction of ATP production both in the enzymatic reaction and in cells treated with this naphthoquinone derivative. MB-6a showed favorable binding to PKM2 in the ATP-bound monomers through docking studies (PDB ID: 4FXF; binding affinity scores ranging from -6.94 to -9.79 kcal/mol) and MD simulations, revealing binding affinities stabilized by key interactions including hydrogen bonds, halogen bonds, and hydrophobic contacts. Conclusions: The findings suggest that MB-6a exerts its antiproliferative activity by disrupting cell glucose metabolism, consequently reducing ATP production and triggering energetic collapse in cancer cells. This study highlights the potential of MB-6a as a lead compound targeting PKM2 and warrants further investigation into its mechanism of action and potential clinical applications.
Collapse
Affiliation(s)
- Lucas Rubini-Dias
- Programa de Pós-Graduação em Ciências Morfológicas, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Fundão, Rio de Janeiro 21941-590, RJ, Brazil; (L.R.-D.); (A.T.A.); (G.O.)
| | - Tácio V. A. Fernandes
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil;
| | - Michele P. de Souza
- Postgraduate Program in Applied Science for Health Products, Faculty of Pharmacy, Fluminense Federal University, Niterói 24020-141, RJ, Brazil;
| | - Déborah Hottz
- Departamento de Ciência Básica, Instituto de Saúde de Nova Fribrugo, Universidade Federal Fluminense, Nova Friburgo 28625-650, RJ, Brazil;
| | - Afonso T. Arruda
- Programa de Pós-Graduação em Ciências Morfológicas, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Fundão, Rio de Janeiro 21941-590, RJ, Brazil; (L.R.-D.); (A.T.A.); (G.O.)
| | - Amanda de A. Borges
- Departamento de Química Orgânica, Instituto de Química, Campus do Valonguinho, Universidade Federal Fluminense, Niterói 24020-150, RJ, Brazil; (A.d.A.B.); (F.d.C.d.S.); (L.d.S.M.F.)
| | - Gabriel Ouverney
- Programa de Pós-Graduação em Ciências Morfológicas, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Fundão, Rio de Janeiro 21941-590, RJ, Brazil; (L.R.-D.); (A.T.A.); (G.O.)
| | - Fernando de C. da Silva
- Departamento de Química Orgânica, Instituto de Química, Campus do Valonguinho, Universidade Federal Fluminense, Niterói 24020-150, RJ, Brazil; (A.d.A.B.); (F.d.C.d.S.); (L.d.S.M.F.)
| | - Luana da S. M. Forezi
- Departamento de Química Orgânica, Instituto de Química, Campus do Valonguinho, Universidade Federal Fluminense, Niterói 24020-150, RJ, Brazil; (A.d.A.B.); (F.d.C.d.S.); (L.d.S.M.F.)
| | | | - Bruno K. Robbs
- Departamento de Ciência Básica, Instituto de Saúde de Nova Fribrugo, Universidade Federal Fluminense, Nova Friburgo 28625-650, RJ, Brazil;
| |
Collapse
|
21
|
Corner TP, Salah E, Tumber A, Kaur S, Nakashima Y, Allen MD, Schnaubelt LI, Fiorini G, Brewitz L, Schofield CJ. Crystallographic and Selectivity Studies on the Approved HIF Prolyl Hydroxylase Inhibitors Desidustat and Enarodustat. ChemMedChem 2024; 19:e202400504. [PMID: 39291299 DOI: 10.1002/cmdc.202400504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/19/2024]
Abstract
Prolyl hydroxylase domain-containing proteins 1-3 (PHD1-3) are 2-oxoglutarate (2OG)-dependent oxygenases catalysing C-4 hydroxylation of prolyl residues in α-subunits of the heterodimeric transcription factor hypoxia-inducible factor (HIF), modifications that promote HIF-α degradation via the ubiquitin-proteasome pathway. Pharmacological inhibition of the PHDs induces HIF-α stabilisation, so promoting HIF target gene transcription. PHD inhibitors are used to treat anaemia caused by chronic kidney disease (CKD) due to their ability to stimulate erythropoietin (EPO) production. We report studies on the effects of the approved PHD inhibitors Desidustat and Enarodustat, and the clinical candidate TP0463518, on activities of a representative set of isolated recombinant human 2OG oxygenases. The three molecules manifest selectivity for PHD inhibition over that of the other 2OG oxygenases evaluated. We obtained crystal structures of Desidustat and Enarodustat in complex with the human 2OG oxygenase factor inhibiting hypoxia-inducible factor-α (FIH), which, together with modelling studies, inform on the binding modes of Desidustat and Enarodustat to active site Fe(II) in 2OG oxygenases, including PHD1-3. The results will help in the design of selective inhibitors of both the PHDs and other 2OG oxygenases, which are of medicinal interest due to their involvement inter alia in metabolic regulation, epigenetic signalling, DNA-damage repair, and agrochemical resistance.
Collapse
Affiliation(s)
- Thomas P Corner
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
- Present Address: Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut, 06511, United States of America
| | - Eidarus Salah
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
| | - Anthony Tumber
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
| | - Samanpreet Kaur
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
| | - Yu Nakashima
- Institute of Natural Medicine, University of Toyama, 2630-Sugitani, Toyama, 930-0194, Japan
| | - Mark D Allen
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
| | - Lara I Schnaubelt
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
| | - Giorgia Fiorini
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
| | - Lennart Brewitz
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
| | - Christopher J Schofield
- Chemistry Research Laboratory, Department of Chemistry and the Ineos Oxford Institute for Antimicrobial Research, University of Oxford, 12 Mansfield Road, Oxford, OX1 3TA, United Kingdom
| |
Collapse
|
22
|
Van R, Pan X, Rostami S, Liu J, Agarwal PK, Brooks B, Rajan R, Shao Y. Exploring CRISPR-Cas9 HNH-Domain-Catalyzed DNA Cleavage Using Accelerated Quantum Mechanical Molecular Mechanical Free Energy Simulation. Biochemistry 2024. [PMID: 39680038 DOI: 10.1021/acs.biochem.4c00651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
The target DNA (tDNA) cleavage catalyzed by the CRISPR Cas9 enzyme is a critical step in the Cas9-based genome editing technologies. Previously, the tDNA cleavage from an active SpyCas9 enzyme conformation was modeled by Palermo and co-workers (Nierzwicki et al., Nat. Catal. 2022 5, 912) using ab initio quantum mechanical molecular mechanical (ai-QM/MM) free energy simulations, where the free energy barrier was found to be more favorable than that from a pseudoactive enzyme conformation. In this work, we performed ai-QM/MM simulations based on another catalytically active conformation (PDB 7Z4J) of the Cas9 HNH domain from cryo-electron microscopy experiments. For the wildtype enzyme, we acquired a free energy profile for the tDNA cleavage that is largely consistent with the previous report. Furthermore, we explored the role of the active-site K866 residue on the catalytic efficiency by modeling the K866A mutant and found that the K866A mutation increased the reaction free energy barrier, which is consistent with the experimentally observed reduction in the enzyme activity.
Collapse
Affiliation(s)
- Richard Van
- Department of Chemistry and Biochemistry, University of Oklahoma, 101 Stephenson Pkwy, Norman, Oklahoma 73019, United States
- Laboratory of Computational Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Xiaoliang Pan
- Department of Chemistry and Biochemistry, University of Oklahoma, 101 Stephenson Pkwy, Norman, Oklahoma 73019, United States
| | - Saadi Rostami
- Department of Chemistry and Biochemistry, University of Oklahoma, 101 Stephenson Pkwy, Norman, Oklahoma 73019, United States
| | - Jin Liu
- Department of Pharmaceutical Sciences, University of North Texas System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, Texas 76107, United States
| | - Pratul K Agarwal
- Department of Physiological Sciences and High Performance Computing Center, Oklahoma State University, 106 Math Sciences, Stillwater, Oklahoma 74078, United States
| | - Bernard Brooks
- Laboratory of Computational Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Rakhi Rajan
- Department of Chemistry and Biochemistry, University of Oklahoma, 101 Stephenson Pkwy, Norman, Oklahoma 73019, United States
| | - Yihan Shao
- Department of Chemistry and Biochemistry, University of Oklahoma, 101 Stephenson Pkwy, Norman, Oklahoma 73019, United States
| |
Collapse
|
23
|
Wozniak S, Janson G, Feig M. Accurate Predictions of Molecular Properties of Proteins via Graph Neural Networks and Transfer Learning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.10.627714. [PMID: 39713395 PMCID: PMC11661272 DOI: 10.1101/2024.12.10.627714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Machine learning has emerged as a promising approach for predicting molecular properties of proteins, as it addresses limitations of experimental and traditional computational methods. Here, we introduce GSnet, a graph neural network (GNN) trained to predict physicochemical and geometric properties including solvation free energies, diffusion constants, and hydrodynamic radii, based on three-dimensional protein structures. By leveraging transfer learning, pre-trained GSnet embeddings were adapted to predict solvent-accessible surface area (SASA) and residue-specific pKa values, achieving high accuracy and generalizability. Notably, GSnet outperformed existing protein embeddings for SASA prediction, and a locally charge-aware variant, aLCnet, approached the accuracy of simulation-based and empirical methods for pKa prediction. Our GNN framework demonstrated robustness across diverse datasets, including intrinsically disordered peptides, and scalability for high-throughput applications. These results highlight the potential of GNN-based embeddings and transfer learning to advance protein structure analysis, providing a foundation for integrating predictive models into proteome-wide studies and structural biology pipelines.
Collapse
Affiliation(s)
- Spencer Wozniak
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Giacomo Janson
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Michael Feig
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
24
|
Benício LFMA, Nascimento ÉCM, Martins JBL. Docking heparan sulfate-based ligands as a promising inhibitor for SARS-CoV-2. J Mol Model 2024; 31:19. [PMID: 39666205 DOI: 10.1007/s00894-024-06236-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024]
Abstract
CONTEXT Heparan sulfate (HS) linear polysaccharide glycosaminoglycan compound is linked to components from the cell surface and the extracellular matrix. HS mediates SARS-CoV-2 infection through spike protein binding to cell surface receptors and is required to bind ACE2, prompting the need for electronic structure and molecular docking evaluation of this core system to exploit this attachment in developing new derivatives. Therefore, we have studied five molecules based on HS using molecular docking and electronic structure analysis. Non-covalent interaction analysis shows hydrogen bonding and van der Waals interactions in the binding to RBD-ACE2 interface and 3CLpro. SDM3 and SDM1 molecules present the lowest gap, including solvent effect under 154.6 kcal/mol, and exhibit the most reactivity behavior in this group, potentially leading to enhanced interaction in docking studies. METHODS Heparan sulfate and four derivatives were optimized using B3LYP functional with two basis sets 6-31 + G(d,p) and def2SVP. Electronic structure was used to explore the main interactions and the reactivity of these molecules, and these optimized structures were used in the molecular docking study against 3CLpro, RBD, and ACE2.
Collapse
Affiliation(s)
- Luiz F M A Benício
- Department of Pharmacy, Faculty of Health Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil
| | - Érica C M Nascimento
- Computational Chemistry Laboratory, Institute of Chemistry, University of Brasilia, Brasilia, DF, 70910-900, Brazil
| | - João B L Martins
- Department of Pharmacy, Faculty of Health Sciences, University of Brasília, Brasília, DF, 70910-900, Brazil.
- Computational Chemistry Laboratory, Institute of Chemistry, University of Brasilia, Brasilia, DF, 70910-900, Brazil.
| |
Collapse
|
25
|
Chang S, Moon R, Nam D, Lee SW, Yoon I, Lee DS, Choi S, Paek E, Hwang D, Hur JK, Nam Y, Chang R, Park H. Hypoxia increases methylated histones to prevent histone clipping and heterochromatin redistribution during Raf-induced senescence. Nucleic Acids Res 2024:gkae1210. [PMID: 39660649 DOI: 10.1093/nar/gkae1210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 11/18/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024] Open
Abstract
Hypoxia enhances histone methylation by inhibiting oxygen- and α-ketoglutarate-dependent demethylases, resulting in increased methylated histones. This study reveals how hypoxia-induced methylation affects histone clipping and the reorganization of heterochromatin into senescence-associated heterochromatin foci (SAHF) during oncogene-induced senescence (OIS) in IMR90 human fibroblasts. Notably, using top-down proteomics, we discovered specific cleavage sites targeted by Cathepsin L (CTSL) in H3, H2B and H4 during Raf activation, identifying novel sites in H2B and H4. Hypoxia counteracts CTSL-mediated histone clipping by promoting methylation without affecting CTSL's activity. This increase in methylation under hypoxia protects against clipping, reshaping the epigenetic landscape and influencing chromatin accessibility, as shown by ATAC-seq analysis. These insights underscore the pivotal role of hypoxia-induced histone methylation in protecting chromatin from significant epigenetic shifts during cellular aging.
Collapse
Affiliation(s)
- Soojeong Chang
- Department of Life Science, University of Seoul, Seoul 02504, Republic of Korea
| | - Ramhee Moon
- Department of Life Science, University of Seoul, Seoul 02504, Republic of Korea
| | - Dowoon Nam
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Sang-Won Lee
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea
| | - Insoo Yoon
- Department of Life Science, University of Seoul, Seoul 02504, Republic of Korea
| | - Dong-Sung Lee
- Department of Life Science, University of Seoul, Seoul 02504, Republic of Korea
| | - Seunghyuk Choi
- Department of Computer Science, Hanyang University, Seoul 04763, Republic of Korea
| | - Eunok Paek
- Department of Computer Science, Hanyang University, Seoul 04763, Republic of Korea
| | - Daehee Hwang
- School of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Junho K Hur
- Department of Genetics, College of Medicine, Hanyang University, Seoul 04763, Republic of Korea
| | - Youhyun Nam
- Department of Applied Chemistry, University of Seoul, Seoul 02504, Republic of Korea
| | - Rakwoo Chang
- Department of Applied Chemistry, University of Seoul, Seoul 02504, Republic of Korea
| | - Hyunsung Park
- Department of Life Science, University of Seoul, Seoul 02504, Republic of Korea
- Department of Applied Chemistry, University of Seoul, Seoul 02504, Republic of Korea
| |
Collapse
|
26
|
Li J, Duran C, Pogrányi B, Cornish KAS, Cartwright J, Osuna S, Unsworth WP, Grogan G. Divergent Oxidation Reactions of E- and Z-Allylic Primary Alcohols by an Unspecific Peroxygenase. Angew Chem Int Ed Engl 2024:e202422241. [PMID: 39655807 DOI: 10.1002/anie.202422241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024]
Abstract
Unspecific peroxygenases (UPOs) catalyze the selective oxygenation of organic substrates using only hydrogen peroxide as the external oxidant. The PaDa-I variant of the UPO from Agrocybe aegerita catalyses the oxidation of Z- and E-allylic alcohols with complementary selectivity, giving epoxide and carboxylic acid/aldehyde products respectively. Both reactions can be performed on preparative scale with isolated yields up to 80 %, and the epoxidations proceed with excellent enantioselectivity (>99 % ee). The divergent reactions can also be used to transform E/Z mixtures of allylic alcohols, enabling both product series to be isolated from a single reaction. The utility of the epoxidation method is exemplified in the total synthesis of both enantiomers of the insect pheromone disparlure, including a highly enantioselective gram-scale transformation. These reactions provide further evidence for the potential of UPOs as catalysts for the scalable preparation of important oxygenated intermediates.
Collapse
Affiliation(s)
- Jiacheng Li
- Department of Chemistry, University of York, Heslington, York, U.K., YO10 5DD
| | - Cristina Duran
- Institut de Química Computacional i Catàlisi and Departament de Química, c/ Maria Aurèlia, Capmany 69, 17003, Girona, Spain
| | - Balázs Pogrányi
- Department of Chemistry, University of York, Heslington, York, U.K., YO10 5DD
| | - Katy A S Cornish
- Department of Chemistry, University of York, Heslington, York, U.K., YO10 5DD
- Department of Biology, University of York, Heslington, York, YO10 5DD U.K
| | - Jared Cartwright
- Department of Biology, University of York, Heslington, York, YO10 5DD U.K
| | - Sílvia Osuna
- Institut de Química Computacional i Catàlisi and Departament de Química, c/ Maria Aurèlia, Capmany 69, 17003, Girona, Spain
- ICREA, Pg. Lluís, Companys 23, 08010, Barcelona, Spain
| | - William P Unsworth
- Department of Chemistry, University of York, Heslington, York, U.K., YO10 5DD
| | - Gideon Grogan
- Department of Chemistry, University of York, Heslington, York, U.K., YO10 5DD
| |
Collapse
|
27
|
Chen Z, Inague A, Kaushal K, Fazeli G, Schilling D, Xavier da Silva TN, Dos Santos AF, Cheytan T, Freitas FP, Yildiz U, Viviani LG, Lima RS, Pinz MP, Medeiros I, Iijima TS, Alegria TGP, Pereira da Silva R, Diniz LR, Weinzweig S, Klein-Seetharaman J, Trumpp A, Mañas A, Hondal R, Bartenhagen C, Fischer M, Shimada BK, Seale LA, Chillon TS, Fabiano M, Schomburg L, Schweizer U, Netto LE, Meotti FC, Dick TP, Alborzinia H, Miyamoto S, Friedmann Angeli JP. PRDX6 contributes to selenocysteine metabolism and ferroptosis resistance. Mol Cell 2024; 84:4645-4659.e9. [PMID: 39547224 DOI: 10.1016/j.molcel.2024.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/02/2024] [Accepted: 10/23/2024] [Indexed: 11/17/2024]
Abstract
Selenocysteine (Sec) metabolism is crucial for cellular function and ferroptosis prevention and begins with the uptake of the Sec carrier, selenoprotein P (SELENOP). Following uptake, Sec released from SELENOP is metabolized via selenocysteine lyase (SCLY), producing selenide, a substrate for selenophosphate synthetase 2 (SEPHS2), which provides the essential selenium donor, selenophosphate (H2SePO3-), for the biosynthesis of the Sec-tRNA. Here, we discovered an alternative pathway in Sec metabolism mediated by peroxiredoxin 6 (PRDX6), independent of SCLY. Mechanistically, we demonstrate that PRDX6 can readily react with selenide and interact with SEPHS2, potentially acting as a selenium delivery system. Moreover, we demonstrate the functional significance of this alternative route in human cancer cells, revealing a notable association between elevated expression of PRDX6 and human MYCN-amplified neuroblastoma subtype. Our study sheds light on a previously unrecognized aspect of Sec metabolism and its implications in ferroptosis, offering further possibilities for therapeutic exploitation.
Collapse
Affiliation(s)
- Zhiyi Chen
- Rudolf Virchow Zentrum (RVZ), Center for Integrative and Translational Bioimaging, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Alex Inague
- Rudolf Virchow Zentrum (RVZ), Center for Integrative and Translational Bioimaging, University of Wuerzburg, 97080 Wuerzburg, Germany; Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil
| | - Kamini Kaushal
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Gholamreza Fazeli
- Rudolf Virchow Zentrum (RVZ), Center for Integrative and Translational Bioimaging, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Danny Schilling
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Thamara N Xavier da Silva
- Rudolf Virchow Zentrum (RVZ), Center for Integrative and Translational Bioimaging, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Ancely Ferreira Dos Santos
- Rudolf Virchow Zentrum (RVZ), Center for Integrative and Translational Bioimaging, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Tasneem Cheytan
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Florencio Porto Freitas
- Rudolf Virchow Zentrum (RVZ), Center for Integrative and Translational Bioimaging, University of Wuerzburg, 97080 Wuerzburg, Germany
| | - Umut Yildiz
- European Molecular Biology Laboratory, Genome Biology Unit, 69117 Heidelberg, Germany
| | - Lucas Gasparello Viviani
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil
| | - Rodrigo Santiago Lima
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil
| | - Mikaela Peglow Pinz
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil
| | - Isadora Medeiros
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil
| | - Thais Satie Iijima
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil
| | - Thiago Geronimo Pires Alegria
- Biosciences Institute, Department of Genetics and Evolutionary Biology, Universidade de São Paulo, 05508900 Sao Paulo, Brazil
| | - Railmara Pereira da Silva
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil
| | - Larissa Regina Diniz
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil
| | - Simon Weinzweig
- School of Molecular Sciences, Arizona State University, Phoenix, AZ 85281, USA
| | | | - Andreas Trumpp
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Adriana Mañas
- Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, IdiPAZ, Hospital Universitario La Paz, 28049 Madrid, Spain; IdiPAZ-CNIO Pediatric Onco-Hematology Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), 28049 Madrid, Spain
| | - Robert Hondal
- Department of Biochemistry, University of Vermont, Burlington, VT 05405, USA
| | - Christoph Bartenhagen
- Center for Molecular Medicine Cologne (CMMC) and Department of Experimental Pediatric Oncology, University Children's Hospital, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Matthias Fischer
- Center for Molecular Medicine Cologne (CMMC) and Department of Experimental Pediatric Oncology, University Children's Hospital, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Briana K Shimada
- Pacific Biosciences Research Center, University of Hawaii at Manoa, Honolulu, HI 96848, USA
| | - Lucia A Seale
- Pacific Biosciences Research Center, University of Hawaii at Manoa, Honolulu, HI 96848, USA
| | - Thilo Samson Chillon
- Institute for Experimental Endocrinology, CVK, Charité-Universtitätsmedizin Berlin, 10115 Berlin, Germany
| | - Marietta Fabiano
- Institut für Biochemie und Molekularbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany
| | - Lutz Schomburg
- Institute for Experimental Endocrinology, CVK, Charité-Universtitätsmedizin Berlin, 10115 Berlin, Germany
| | - Ulrich Schweizer
- Institut für Biochemie und Molekularbiologie, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany
| | - Luis E Netto
- Biosciences Institute, Department of Genetics and Evolutionary Biology, Universidade de São Paulo, 05508900 Sao Paulo, Brazil
| | - Flavia C Meotti
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil
| | - Tobias P Dick
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Hamed Alborzinia
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Sayuri Miyamoto
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, 05508000 Sao Paulo, Brazil.
| | - José Pedro Friedmann Angeli
- Rudolf Virchow Zentrum (RVZ), Center for Integrative and Translational Bioimaging, University of Wuerzburg, 97080 Wuerzburg, Germany.
| |
Collapse
|
28
|
Wu J, Jonniya NA, Hirakis SP, Olivieri C, Veglia G, Kornev AP, Taylor SS. Role of the αC-β4 loop in protein kinase structure and dynamics. eLife 2024; 12:RP91980. [PMID: 39630082 PMCID: PMC11616992 DOI: 10.7554/elife.91980] [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] [Indexed: 12/07/2024] Open
Abstract
Although the αC-β4 loop is a stable feature of all protein kinases, the importance of this motif as a conserved element of secondary structure, as well as its links to the hydrophobic architecture of the kinase core, has been underappreciated. We first review the motif and then describe how it is linked to the hydrophobic spine architecture of the kinase core, which we first discovered using a computational tool, local spatial Pattern (LSP) alignment. Based on NMR predictions that a mutation in this motif abolishes the synergistic high-affinity binding of ATP and a pseudo substrate inhibitor, we used LSP to interrogate the F100A mutant. This comparison highlights the importance of the αC-β4 loop and key residues at the interface between the N- and C-lobes. In addition, we delved more deeply into the structure of the apo C-subunit, which lacks ATP. While apo C-subunit showed no significant changes in backbone dynamics of the αC-β4 loop, we found significant differences in the side chain dynamics of K105. The LSP analysis suggests disruption of communication between the N- and C-lobes in the F100A mutant, which would be consistent with the structural changes predicted by the NMR spectroscopy.
Collapse
Affiliation(s)
- Jian Wu
- Department of Pharmacology, University of California, San DiegoSan DiegoUnited States
| | - Nisha A Jonniya
- Department of Pharmacology, University of California, San DiegoSan DiegoUnited States
| | - Sophia P Hirakis
- Department of Chemistry and Biochemistry, University of California, San DiegoSan DiegoUnited States
| | - Cristina Olivieri
- Department of Biochemistry, Molecular Biology, and Biophysics, University of MinnesotaMinneapolisUnited States
| | - Gianluigi Veglia
- Department of Biochemistry, Molecular Biology, and Biophysics, University of MinnesotaMinneapolisUnited States
- Department of Chemistry, University of MinnesotaMinneapolisUnited States
| | - Alexandr P Kornev
- Department of Pharmacology, University of California, San DiegoSan DiegoUnited States
| | - Susan S Taylor
- Department of Pharmacology, University of California, San DiegoSan DiegoUnited States
- Department of Chemistry and Biochemistry, University of California, San DiegoSan DiegoUnited States
| |
Collapse
|
29
|
Coricello A, Nardone AJ, Lupia A, Gratteri C, Vos M, Chaptal V, Alcaro S, Zhu W, Takagi Y, Richards NGJ. 3D variability analysis reveals a hidden conformational change controlling ammonia transport in human asparagine synthetase. Nat Commun 2024; 15:10538. [PMID: 39627226 PMCID: PMC11615228 DOI: 10.1038/s41467-024-54912-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 11/20/2024] [Indexed: 12/06/2024] Open
Abstract
Advances in X-ray crystallography and cryogenic electron microscopy (cryo-EM) offer the promise of elucidating functionally relevant conformational changes that are not easily studied by other biophysical methods. Here we show that 3D variability analysis (3DVA) of the cryo-EM map for wild-type (WT) human asparagine synthetase (ASNS) identifies a functional role for the Arg-142 side chain and test this hypothesis experimentally by characterizing the R142I variant in which Arg-142 is replaced by isoleucine. Support for Arg-142 playing a role in the intramolecular translocation of ammonia between the active site of the enzyme is provided by the glutamine-dependent synthetase activity of the R142 variant relative to WT ASNS, and MD simulations provide a possible molecular mechanism for these findings. Combining 3DVA with MD simulations is a generally applicable approach to generate testable hypotheses of how conformational changes in buried side chains might regulate function in enzymes.
Collapse
Affiliation(s)
- Adriana Coricello
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Catanzaro, Italy
- Dipartimento di Scienze Biomolecolari, Università degli Studi di Urbino "Carlo Bo", Urbino, Italy
| | - Alanya J Nardone
- Department of Chemistry & Biochemistry, Florida State University, Tallahassee, FL, USA
| | - Antonio Lupia
- Net4Science Academic Spin-Off, Università "Magna Græcia" di Catanzaro, Catanzaro, Italy
- Dipartimento di Scienze della vita e dell'ambiente, Università degli Studi di Cagliari, Cagliari, Italy
| | - Carmen Gratteri
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Catanzaro, Italy
| | - Matthijn Vos
- NanoImaging Core Facility, Centre de Resources et Recherches Technologiques, Institut Pasteur, Paris, France
| | - Vincent Chaptal
- Molecular Microbiology and Structural Biochemistry Laboratory, CNRS UMR 5086, University of Lyon, Lyon, France
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università "Magna Græcia" di Catanzaro, Catanzaro, Italy.
- Net4Science Academic Spin-Off, Università "Magna Græcia" di Catanzaro, Catanzaro, Italy.
| | - Wen Zhu
- Department of Chemistry & Biochemistry, Florida State University, Tallahassee, FL, USA.
| | - Yuichiro Takagi
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Nigel G J Richards
- School of Chemistry, Cardiff University, Park Place, Cardiff, UK.
- Foundation for Applied Molecular Evolution, Alachua, FL, USA.
| |
Collapse
|
30
|
Ma S, Li M, Yan S, Wang Y, Wang B, Luo W. Enhancing the specific activity of 3α-hydroxysteroid dehydrogenase through cross-regional combinatorial mutagenesis. Int J Biol Macromol 2024; 283:137014. [PMID: 39486711 DOI: 10.1016/j.ijbiomac.2024.137014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/20/2024] [Accepted: 10/26/2024] [Indexed: 11/04/2024]
Abstract
3α-Hydroxysteroid dehydrogenase (3α-HSD) from Comamonas testosteroni is widely used in clinical settings to measure serum total bile acid levels. However, its low enzymatic activity leads to high operational costs. In this study, we employed a combinatorial mutagenesis approach to systematically identify potential key mutation sites within the enzyme. The enzyme molecule was segmented into distinct regions, and a comprehensive strategy integrating substrate pocket engineering, binding energy calculations, and deep learning techniques was used. Through experimental verification, single-point mutants from the mutation library with enhanced enzymatic activity by at least 1.5-fold were identified. Through iterative combinatorial mutations of them, the optimal mutant H119A/R201G/R216L was obtained. This mutant exhibited a specific activity of 34.18 U/mg towards deoxycholic acid, representing a 6.85-fold increase over the wild-type (WT) enzyme. Additionally, the optimal temperature of the mutant increased from 35 °C to 40 °C, and its turnover number and catalytic efficiency increased by 6.4-fold and 9.4-fold, respectively. Quantum mechanics/molecular mechanics (QM/MM) calculations indicated that the energy barrier of the dehydrogenase reaction was reduced in the H119A/R201G/R216L mutant compared to that of the WT enzyme. Specifically, the R201G mutation significantly reduced the electric field strength along the 3α-hydroxyl group, facilitating its deprotonation. This study provides insights into enhancing enzymatic efficiency through strategic mutagenesis and elucidates mechanistic changes that optimize enzyme performance for clinical and biotechnological applications.
Collapse
Affiliation(s)
- Siqi Ma
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China
| | - Musen Li
- State Key Laboratory of Physical Chemistry of Solid Surfaces and Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Shengheng Yan
- State Key Laboratory of Physical Chemistry of Solid Surfaces and Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yi Wang
- Department of Biological and Agricultural Engineering, University of California, Davis, CA, 95616, USA
| | - Binju Wang
- State Key Laboratory of Physical Chemistry of Solid Surfaces and Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| | - Wei Luo
- The Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
31
|
Kotzampasi DM, Papadourakis M, Burke JE, Cournia Z. Free energy landscape of the PI3Kα C-terminal activation. Comput Struct Biotechnol J 2024; 23:3118-3131. [PMID: 39229338 PMCID: PMC11369385 DOI: 10.1016/j.csbj.2024.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/05/2024] [Accepted: 07/05/2024] [Indexed: 09/05/2024] Open
Abstract
The gene PIK3CA, encoding the catalytic subunit p110α of PI3Kα, is the second most frequently mutated gene in cancer, with the highest frequency oncogenic mutants occurring in the C-terminus of the kinase domain. The C-terminus has a dual function in regulating the kinase, playing a putative auto-inhibitory role for kinase activity and being absolutely essential for binding to the cell membrane. However, the molecular mechanisms by which these C-terminal oncogenic mutations cause PI3Kα overactivation remain unclear. To understand how a spectrum of C-terminal mutations of PI3Kα alter kinase activity compared to the WT, we perform unbiased and biased Molecular Dynamics simulations of several C-terminal mutants and report the free energy landscapes for the C-terminal "closed-to-open" transition in the WT, H1047R, G1049R, M1043L and N1068KLKR mutants. Results are consistent with HDX-MS experimental data and provide a molecular explanation why H1047R and G1049R reorient the C-terminus with a different mechanism compared to the WT and M1043L and N1068KLKR mutants. Moreover, we show that in the H1047R mutant, the cavity, where the allosteric ligands STX-478 and RLY-2608 bind, is more accessible contrary to the WT. This study provides insights into the molecular mechanisms underlying activation of oncogenic PI3Kα by C-terminal mutations and represents a valuable resource for continued efforts in the development of mutant selective inhibitors as therapeutics.
Collapse
Affiliation(s)
- Danai Maria Kotzampasi
- Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
- Department of Biology, University of Crete, Heraklion 71500, Greece
| | | | - John E. Burke
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC V8W 2Y2, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Zoe Cournia
- Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece
| |
Collapse
|
32
|
Rollins ZA, Curtis MB, George SC, Faller R. A Computational Strategy for the Rapid Identification and Ranking of Patient-Specific T Cell Receptors Bound to Neoantigens. Macromol Rapid Commun 2024; 45:e2400225. [PMID: 38839076 PMCID: PMC11661661 DOI: 10.1002/marc.202400225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/02/2024] [Indexed: 06/07/2024]
Abstract
T cell receptor (TCR) recognition of a peptide-major histocompatibility complex (pMHC) is crucial for adaptive immune response. The identification of therapeutically relevant TCR-pMHC protein pairs is a bottleneck in the implementation of TCR-based immunotherapies. The ability to computationally design TCRs to target a specific pMHC requires automated integration of next-generation sequencing, protein-protein structure prediction, molecular dynamics, and TCR ranking. A pipeline to evaluate patient-specific, sequence-based TCRs to a target pMHC is presented. Using the three most frequently expressed TCRs from 16 colorectal cancer patients, the protein-protein structure of the TCRs to the target CEA peptide-MHC is predicted using Modeller and ColabFold. TCR-pMHC structures are compared using automated equilibration and successive analysis. ColabFold generated configurations require an ≈2.5× reduction in equilibration time of TCR-pMHC structures compared to Modeller. The structural differences between Modeller and ColabFold are demonstrated by root mean square deviation (≈0.20 nm) between clusters of equilibrated configurations, which impact the number of hydrogen bonds and Lennard-Jones contacts between the TCR and pMHC. TCR ranking criteria that may prioritize TCRs for evaluation of in vitro immunogenicity are identified, and this ranking is validated by comparing to state-of-the-art machine learning-based methods trained to predict the probability of TCR-pMHC binding.
Collapse
Affiliation(s)
- Zachary A. Rollins
- Department of Chemical EngineeringUniversity of CaliforniaDavis, 1 Shields Ave, Bainer HallDavisCA95616USA
| | - Matthew B. Curtis
- Department of Biomedical EngineeringUniversity of CaliforniaDavis, 451 E. Health Sciences Dr., GBSF 2303DavisCA95616USA
| | - Steven C. George
- Department of Biomedical EngineeringUniversity of CaliforniaDavis, 451 E. Health Sciences Dr., GBSF 2303DavisCA95616USA
| | - Roland Faller
- Department of Chemical EngineeringUniversity of CaliforniaDavis, 1 Shields Ave, Bainer HallDavisCA95616USA
- Department of Chemical EngineeringTexas Tech UniversityLubbockTX79409USA
| |
Collapse
|
33
|
Sankaran SV, Saiba R, Sikdar S, Vemparala S. Correlation Between Antimicrobial Structural Classes and Membrane Partitioning: Role of Emerging Lipid Packing Defects. J Membr Biol 2024; 257:307-321. [PMID: 39037449 PMCID: PMC11584508 DOI: 10.1007/s00232-024-00318-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024]
Abstract
In this study, a combination of bioinformatics and molecular dynamics simulations is employed to investigate the partitioning behavior of different classes of antimicrobial peptides (AMPs) into model membranes. The main objective is to identify any correlations between the structural characteristics of AMPs and their membrane identification and early-stage partitioning mechanisms. The simulation results reveal distinct membrane interactions among the various structural classes of AMPs, particularly in relation to the generation and subsequent interaction with lipid packing defects. Notably, AMPs with a structure-less coil conformation generate a higher number of deep and shallow defects, which are larger in size compared to other classes of AMPs. AMPs with helical component demonstrated the deepest insertion into the membrane. On the other hand, AMPs with a significant percentage of beta sheets tend to adsorb onto the membrane surface, suggesting a potentially distinct partitioning mechanism attributed to their structural rigidity. These findings highlight the diverse membrane interactions and partitioning mechanisms exhibited by different structural classes of AMPs.
Collapse
Affiliation(s)
- S V Sankaran
- The Institute of Mathematical Sciences, C.I.T. Campus, Taramani, Chennai, Tamil Nadu, 600113, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, 400094, India
| | - Roni Saiba
- The Institute of Mathematical Sciences, C.I.T. Campus, Taramani, Chennai, Tamil Nadu, 600113, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, 400094, India
| | - Samapan Sikdar
- The Institute of Mathematical Sciences, C.I.T. Campus, Taramani, Chennai, Tamil Nadu, 600113, India.
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, 400094, India.
| | - Satyavani Vemparala
- The Institute of Mathematical Sciences, C.I.T. Campus, Taramani, Chennai, Tamil Nadu, 600113, India.
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, Maharashtra, 400094, India.
| |
Collapse
|
34
|
Hong R, Alagbe BD, Mattei A, Sheikh AY, Tuckerman ME. Enhanced and Efficient Predictions of Dynamic Ionization through Constant-pH Adiabatic Free Energy Dynamics. J Chem Theory Comput 2024; 20:10010-10021. [PMID: 39513519 PMCID: PMC11603612 DOI: 10.1021/acs.jctc.4c00704] [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: 05/31/2024] [Revised: 10/14/2024] [Accepted: 10/16/2024] [Indexed: 11/15/2024]
Abstract
Dynamic or structurally induced ionization is a critical aspect of many physical, chemical, and biological processes. Molecular dynamics (MD) based simulation approaches, specifically constant pH MD methods, have been developed to simulate ionization states of molecules or proteins under experimentally or physiologically relevant conditions. While such approaches are now widely utilized to predict ionization sites of macromolecules or to study physical or biological phenomena, they are often computationally expensive and require long simulation times to converge. In this article, using the principles of adiabatic free energy dynamics, we introduce an efficient technique for performing constant pH MD simulations within the framework of the adiabatic free energy dynamics (AFED) approach. We call the new approach pH-AFED. We show that pH-AFED provides highly accurate predictions of protein residue pKa values, with a MUE of 0.5 pKa units when coupled with driven adiabatic free energy dynamics (d-AFED), while reducing the required simulation times by more than an order of magnitude. In addition, pH-AFED can be easily integrated into most constant pH MD codes or implementations and flexibly adapted to work in conjunction with enhanced sampling algorithms that target collective variables. We demonstrate that our approaches, with both pH-AFED standalone as well as pH-AFED combined with collective variable based enhanced sampling, provide promising predictive accuracy, with a MUE of 0.6 and 0.5 pKa units respectively, on a diverse range of proteins and enzymes, ranging up to 186 residues and 21 titratable sites. Lastly, we demonstrate how this approach can be utilized to understand the in vivo performance engineered antibodies for immunotherapy.
Collapse
Affiliation(s)
- Richard
S. Hong
- AbbVie
Inc., Molecular Profiling and Drug Delivery, Research & Development, 1 N Waukegan Road, North Chicago, Illinois 60064, United States
- Department
of Chemistry, New York University, New York City, New York 10003, United States
| | - Busayo D. Alagbe
- AbbVie
Inc., Molecular Profiling and Drug Delivery, Research & Development, 1 N Waukegan Road, North Chicago, Illinois 60064, United States
| | - Alessandra Mattei
- AbbVie
Inc., Molecular Profiling and Drug Delivery, Research & Development, 1 N Waukegan Road, North Chicago, Illinois 60064, United States
| | - Ahmad Y. Sheikh
- AbbVie
Inc., Molecular Profiling and Drug Delivery, Research & Development, 1 N Waukegan Road, North Chicago, Illinois 60064, United States
| | - Mark E. Tuckerman
- Department
of Chemistry, New York University, New York City, New York 10003, United States
- Courant
Institute of Mathematical Sciences, New
York University, New York, New York 10012, United States
- NYU-ECNU
Center for Computational Chemistry at NYU Shanghai, 3663 Zhongshan Road North, Shanghai 200062, China
- Simons
Center for Computational Physical Chemistry at New York University, New York, New York 10003, United States
| |
Collapse
|
35
|
Brcko IC, de Souza VC, Ribeiro G, Lima ARJ, Martins AJ, Barros CRDS, de Carvalho E, Pereira JS, de Lima LPO, Viala VL, Kashima S, de La Roque DGL, Santos EV, Rodrigues ES, Nunes JA, Torres LS, Caldeira LAV, Palmieri M, Medina CG, de Arruda RA, Lopes RB, Sobrinho GR, Jorge DMDM, Arruda E, Mendes ECBDS, Santos HDO, de Mello ALES, Pereira FM, Gómez MKA, Nardy VB, Henrique B, Vieira LL, Roll MM, de Oliveira EC, Almeida JDPC, da Silva SF, Borges GAL, Furtado KCDL, da Costa PMSSB, Chagas SMDS, Kallás EG, Larh D, Giovanetti M, Nanev Slavov S, Coccuzzo Sampaio S, Elias MC. Comprehensive molecular epidemiology of influenza viruses in Brazil: insights from a nationwide analysis. Virus Evol 2024; 11:veae102. [PMID: 39802823 PMCID: PMC11711486 DOI: 10.1093/ve/veae102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/25/2024] [Accepted: 11/22/2024] [Indexed: 01/16/2025] Open
Abstract
Influenza A and B viruses represent significant global health threats, contributing substantially to morbidity and mortality rates. However, a comprehensive understanding of the molecular epidemiology of these viruses in Brazil, a continental-size country and a crucial hub for the entry, circulation, and dissemination of influenza viruses within South America, still needs to be improved. This study addresses this gap by consolidating data and samples across all Brazilian macroregions, as part of the Center for Viral Surveillance and Serological Assessment project, together with an extensive number of other Brazilian sequences provided by a public database during the epidemic seasons spanning 2021-23. Phylogenetic analysis of the hemagglutinin segment of influenza A/H1N1pdm09, A/H3N2, and influenza B/Victoria-lineage viruses revealed that in 2021 and in the first semester of 2022, the A/H3N2 2a.3 strain was the predominant circulating strain. Subsequently, the A/H3N2 2b became the prevalent strain until October, when it was substituted by A/H1N1pdm09 5a.2a and 5a.2a.1 lineages. This scenario was maintained during the year of 2023. B/Victoria emerged and circulated at low levels between December 2021 and September 2022 and then became coprevalent with A/H1N1pdm09 5a.2a and 5a.2a.1 lineages. The comparison between the vaccine strain A/Darwin/9/2021 and circulating viruses revealed shared mutations to aspartic acid at residues 186 and 225 across all A/H3N2 lineages from 2021 to 2023, altering the charge in the receptor-binding domain. For A/H1N1pdm09, the 2022 consensus of 5a.2a.1 and the vaccine strain A/Victoria/2570/2019 showed 14 amino acid substitutions. Key residues H180, D187, K219, R223, E224, and T133 are involved in hydrogen interactions with sialic acids, while N130, K142, and D222 may contribute to distance interactions based on docking analyses. Importantly, distinct influenza A lineage frequency patterns were observed across Brazil's macroregions, underscoring the regional variations in virus circulation. This study characterizes influenza A and B viruses circulating in Brazil, providing insights into their circulation patterns and dynamics across Brazilian macroregions. These findings hold significant implications for public health interventions, informing strategies to mitigate transmission risks, optimize vaccination efforts, and enhance outbreak control measures.
Collapse
Affiliation(s)
- Isabela Carvalho Brcko
- Center for Viral Surveillance and Serological Assessment (CeVIVAS), Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
- Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
| | - Vinicius Carius de Souza
- Center for Viral Surveillance and Serological Assessment (CeVIVAS), Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
- Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
| | - Gabriela Ribeiro
- Center for Viral Surveillance and Serological Assessment (CeVIVAS), Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
- Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
| | - Alex Ranieri Jeronimo Lima
- Center for Viral Surveillance and Serological Assessment (CeVIVAS), Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
- Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
| | - Antonio Jorge Martins
- Center for Viral Surveillance and Serological Assessment (CeVIVAS), Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
- Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
| | - Claudia Renata dos Santos Barros
- Center for Viral Surveillance and Serological Assessment (CeVIVAS), Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
- Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
| | - Eneas de Carvalho
- Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
| | - James Siqueira Pereira
- Center for Viral Surveillance and Serological Assessment (CeVIVAS), Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
- Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
| | - Loyze Paola Oliveira de Lima
- Center for Viral Surveillance and Serological Assessment (CeVIVAS), Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
- Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
| | - Vincent Louis Viala
- Center for Viral Surveillance and Serological Assessment (CeVIVAS), Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
- Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
| | - Simone Kashima
- Center for Viral Surveillance and Serological Assessment (CeVIVAS), Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
- Hemocentro Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, São Paulo 14051-140, Brazil
| | | | - Elaine Vieira Santos
- Hemocentro Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, São Paulo 14051-140, Brazil
| | - Evandra Strazza Rodrigues
- Hemocentro Ribeirão Preto, Rua Tenente Catão Roxo, 2501, Ribeirão Preto, São Paulo 14051-140, Brazil
| | - Juliana Almeida Nunes
- Coordenadoria de Vigilância em Saúde (COVISA), Secretaria Municipal de Saúde (SMS/SP), Prefeitura São Paulo, Rua Siqueira Campos, 176, São Paulo, São Paulo 01509-020, Brazil
| | - Leandro Spalato Torres
- Coordenadoria de Vigilância em Saúde (COVISA), Secretaria Municipal de Saúde (SMS/SP), Prefeitura São Paulo, Rua Siqueira Campos, 176, São Paulo, São Paulo 01509-020, Brazil
| | - Luiz Artur Vieira Caldeira
- Coordenadoria de Vigilância em Saúde (COVISA), Secretaria Municipal de Saúde (SMS/SP), Prefeitura São Paulo, Rua Siqueira Campos, 176, São Paulo, São Paulo 01509-020, Brazil
| | - Melissa Palmieri
- Coordenadoria de Vigilância em Saúde (COVISA), Secretaria Municipal de Saúde (SMS/SP), Prefeitura São Paulo, Rua Siqueira Campos, 176, São Paulo, São Paulo 01509-020, Brazil
| | - Caio Genovez Medina
- Departamento de Atenção Hospitalar de São Bernardo do Campo, Prefeitura São Bernardo do Campo, Rua João Pessoa, 59, São Bernardo do Campo, São Paulo 09715-000, Brazil
| | - Raphael Augusto de Arruda
- Departamento de Atenção Hospitalar de São Bernardo do Campo, Prefeitura São Bernardo do Campo, Rua João Pessoa, 59, São Bernardo do Campo, São Paulo 09715-000, Brazil
| | - Renata Beividas Lopes
- Departamento de Atenção Hospitalar de São Bernardo do Campo, Prefeitura São Bernardo do Campo, Rua João Pessoa, 59, São Bernardo do Campo, São Paulo 09715-000, Brazil
| | - Geraldo Reple Sobrinho
- Secretaria de Saúde de São Bernardo do Campo, Prefeitura São Bernardo do Campo, Rua João Pessoa, 59, São Bernardo do Campo, São Paulo 09715-000, Brazil
| | - Daniel Macedo de Melo Jorge
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Avenida dos Bandeirantes, 3900, Ribeirão Preto, São Paulo 14048-900, Brazil
| | - Eurico Arruda
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Avenida dos Bandeirantes, 3900, Ribeirão Preto, São Paulo 14048-900, Brazil
| | | | - Hazerral de Oliveira Santos
- Laboratório Central de Saúde Pública do Estado de Alagoas (LACEN-AL), Rua Doutor Ernesto Gomes Maranhão, 1773, Maceió, Alagoas 57036-860, Brazil
| | - Arabela Leal e Silva de Mello
- Laboratório Central de Saúde Pública do Estado da Bahia (LACEN-BA), 4ª Avenida, 400, Salvador, Bahia 41745-900, Brazil
| | - Felicidade Mota Pereira
- Laboratório Central de Saúde Pública do Estado da Bahia (LACEN-BA), 4ª Avenida, 400, Salvador, Bahia 41745-900, Brazil
| | - Marcela Kelly Astete Gómez
- Laboratório Central de Saúde Pública do Estado da Bahia (LACEN-BA), 4ª Avenida, 400, Salvador, Bahia 41745-900, Brazil
| | - Vanessa Brandão Nardy
- Laboratório Central de Saúde Pública do Estado da Bahia (LACEN-BA), 4ª Avenida, 400, Salvador, Bahia 41745-900, Brazil
| | - Brenno Henrique
- Laboratório Central de Saúde Pública do Distrito Federal (LACEN-DF), Lotes O e P, Sgan 601, Brasília, Distrito Federal 70.830-010, Brazil
| | - Lucas Luiz Vieira
- Laboratório Central de Saúde Pública do Distrito Federal (LACEN-DF), Lotes O e P, Sgan 601, Brasília, Distrito Federal 70.830-010, Brazil
| | - Mariana Matos Roll
- Laboratório Central de Saúde Pública do Distrito Federal (LACEN-DF), Lotes O e P, Sgan 601, Brasília, Distrito Federal 70.830-010, Brazil
| | - Elaine Cristina de Oliveira
- Laboratório Central de Saúde Pública do Estado de Mato Grosso (LACEN-MT), Rua Santiago, 70, Cuiabá, Mato Grosso 78.060-628, Brazil
| | | | - Stephanni Figueiredo da Silva
- Laboratório Central de Saúde Pública do Estado de Mato Grosso (LACEN-MT), Rua Santiago, 70, Cuiabá, Mato Grosso 78.060-628, Brazil
| | - Gleissy Adriane Lima Borges
- Laboratório Central de Saúde Pública do Estado do Pará (LACEN-PA), Rodovia Augusto Montenegro, 524, Belém, Pará 66823-010, Brazil
| | - Katia Cristina de Lima Furtado
- Laboratório Central de Saúde Pública do Estado do Pará (LACEN-PA), Rodovia Augusto Montenegro, 524, Belém, Pará 66823-010, Brazil
| | | | - Shirley Moreira da Silva Chagas
- Laboratório Central de Saúde Pública do Estado do Pará (LACEN-PA), Rodovia Augusto Montenegro, 524, Belém, Pará 66823-010, Brazil
| | - Esper G Kallás
- Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Rua Doutor Ovídio Pires de Campos, 225, São Paulo, São Paulo 05403-010, Brazil
| | - Daniel Larh
- Instituto de Biociências, Universidade de São Paulo, Rua do Matão, 321, São Paulo, São Paulo 05508-090, Brazil
| | - Marta Giovanetti
- Department of Science and Technology for Humans and the Environment, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, Rome 00128 Rome, Italy
- Instituto Rene Rachou, Fundação Oswaldo Cruz, Avenida Augusto de Lima, 1715, Belo Horizonte, Minas Gerais 30190-002, Brazil
- Climate Amplified Diseases and Epidemics (CLIMADE), CERI, Tygerberg Medical Campus, Cape Town, South Africa & Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Svetoslav Nanev Slavov
- Center for Viral Surveillance and Serological Assessment (CeVIVAS), Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
- Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
| | - Sandra Coccuzzo Sampaio
- Center for Viral Surveillance and Serological Assessment (CeVIVAS), Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
- Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
| | - Maria Carolina Elias
- Center for Viral Surveillance and Serological Assessment (CeVIVAS), Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
- Instituto Butantan, Avenida Vital Brasil, 1500, Butantã, São Paulo, São Paulo 05503-900, Brazil
| |
Collapse
|
36
|
Barbosa ED, Ma Y, Clift HE, Olson LJ, Zhu L, Liu W. Structural Insights into Dopamine Receptor-Ligand Interactions: From Agonists to Antagonists. ACS Chem Neurosci 2024; 15:4123-4131. [PMID: 39485723 DOI: 10.1021/acschemneuro.4c00295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024] Open
Abstract
This study explores the intricacies of dopamine receptor-ligand interactions, focusing on the D1R and D5R subtypes. Using molecular modeling techniques, we investigated the binding of the pan-agonist rotigotine, revealing a universal binding mode at the orthosteric binding pocket. Additionally, we analyze the stability of antagonist-receptor complexes with SKF83566 and SCH23390. By examining the impact of specific mutations on ligand-receptor interactions through computational simulations and thermostability assays, we gain insights into binding stability. Our research also delves into the structural and energetic aspects of antagonist binding to D1R and D5R in their inactive states. These findings enhance our understanding of dopamine receptor pharmacology and hold promise for drug development in central nervous system disorders, opening doors to future research and innovation in this field.
Collapse
MESH Headings
- Dopamine Agonists/pharmacology
- Dopamine Agonists/chemistry
- Humans
- Ligands
- Dopamine Antagonists/pharmacology
- Dopamine Antagonists/chemistry
- Receptors, Dopamine D1/metabolism
- Receptors, Dopamine D1/agonists
- Tetrahydronaphthalenes/pharmacology
- Tetrahydronaphthalenes/chemistry
- Receptors, Dopamine D5/agonists
- Receptors, Dopamine D5/metabolism
- Thiophenes/pharmacology
- Thiophenes/chemistry
- Protein Binding/physiology
- Binding Sites
- Benzazepines/pharmacology
- Benzazepines/chemistry
- Models, Molecular
- 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology
- 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/analogs & derivatives
Collapse
Affiliation(s)
- Emmanuel D Barbosa
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Yuanyuan Ma
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Heather E Clift
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Linda J Olson
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Lan Zhu
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Wei Liu
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| |
Collapse
|
37
|
Beyens O, Corthaut S, Lambeir AM, Van Der Veken P, Sterckx YGJ, De Meester I, De Winter H. An Interdisciplinary Approach Provides Insights into the Pronounced Selectivity of Compound 42 for DPP9. ChemMedChem 2024:e202400700. [PMID: 39552560 DOI: 10.1002/cmdc.202400700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/19/2024]
Abstract
Dipeptidyl peptidase 8 (DPP8) and 9 (DPP9) are proteases gaining significant attention for their role in health and disease. Distinctive studies of these proteases are hampered by their close homology. Furthermore, designing selective compounds is a major challenge due to the highly conserved catalytic site. Here, we provide mechanistic insights underlying the DPP9-over-DPP8 selectivity of the semi-selective inhibitor "Compound 42". We performed enhanced sampling molecular dynamics simulations to investigate the binding pose of "Compound 42", which enabled the design of various DPP9 mutants that were characterized through a combination of biochemical (Ki determinations) and in silico approaches. Our findings show that DPP9 residue F253 is an important selectivity-determining factor. This work marks the discovery and validation of a structural feature that can be exploited for the design of DPP8 or DPP9 selective inhibitors.
Collapse
Affiliation(s)
- Olivier Beyens
- Laboratory of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Sam Corthaut
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Anne-Marie Lambeir
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Pieter Van Der Veken
- Laboratory of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Yann G-J Sterckx
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Ingrid De Meester
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
| | - Hans De Winter
- Laboratory of Medicinal Chemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610, Antwerp, Belgium
| |
Collapse
|
38
|
Mitra A, Pandijothi V, Paul S. Computational insight into the peptide-based inhibition of α-cobratoxin. Phys Chem Chem Phys 2024; 26:28274-28287. [PMID: 39499553 DOI: 10.1039/d4cp03408b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
Snakebite envenoming results in the death of thousands of people each year and has been classified as a neglected tropical disease by the World Health Organization (WHO). The toxins released into the bloodstream of the victim bind to the nicotinic acetylcholine receptor and restrict transmission of nerve impulses leading to paralysis and cardiac arrest. Conventional antibody-based treatments often have adverse side effects or are difficult to perform. Hence, efforts are underway to devise alternative forms of treatment that address these therapeutic shortcomings. Peptide-based inhibitors have recently gained attention due to their high specificity and ease of preparation. Here, we explore the mechanism of a peptide inhibitor of α-cobratoxin using all-atom molecular dynamics (MD) simulations. We also quantify the energetics of the toxin-peptide dissociation process using the non-equilibrium steered MD technique. Our study reveals that the inhibitor migrates close to Loop-II of α-cobratoxin and alters its dimerization tendency. From energy studies, we found that the peptide first binds to one unit of α-cobratoxin in a particular orientation, followed by the binding of a second toxin molecule, which effectively masks the residues that interact with the nicotinic acetylcholine receptor. Our work provides atomic-level insight into the inhibition process that can be utilized in the future design of inhibitors with superior binding capabilities.
Collapse
Affiliation(s)
- Aritra Mitra
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam 781039, India.
| | - Viswas Pandijothi
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam 781039, India.
| | - Sandip Paul
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam 781039, India.
| |
Collapse
|
39
|
Berselli A, Menziani MC, Muniz-Miranda F. Structure and Energetics of PET-Hydrolyzing Enzyme Complexes: A Systematic Comparison from Molecular Dynamics Simulations. J Chem Inf Model 2024; 64:8236-8257. [PMID: 39432831 DOI: 10.1021/acs.jcim.4c01369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Discovered in 2016, the enzyme PETase, secreted by bacterial Ideonella Sakaiensis 201-F6, has an excellent hydrolytic activity toward poly(ethylene terephthalate) (PET) at room temperature, while it decreases at higher temperatures due to the low thermostability. Many variants have been engineered to overcome this limitation, which hinders industrial application. In this work, we systematically compare PETase wild-type (WT) and four mutants (DuraPETase, ThermoPETase, FastPETase, and HotPETase) using standard molecular dynamics (MD) simulations and unbinding free energy calculations. In particular, we analyze the enzymes' structural characteristics and binding to a tetrameric PET chain (PET4) under two temperature conditions: T1─300 K and T2─350 K. Our results indicate that (i) PET4 forms stable complexes with the five enzymes at room temperature (∼300 K) and (ii) most of the interactions are localized close to the active site of the protein, where the W185 and Y87 residues interact with the aromatic rings of the substrate. Specifically, (iii) the W185 side-chain explores different conformations in each variant (a phenomenon known in the literature as "W185 wobbling"). This suggests that the binding pocket retains structural plasticity and flexibility among the variants, facilitating substrate recognition and localization events at moderate temperatures. Moreover, (iv) PET4 establishes aromatic interactions with the catalytic H237 residue, stabilizing the catalytic triad composed of residues S160-H237-D206, and helping the system achieve an effective configuration for the hydrolysis reaction. Conversely, (v) the binding affinity decreases at a higher temperature (∼350 K), retaining moderate interactions only for HotPETase. Finally, (vi) MD simulations of complexes formed with poly(ethylene-2,5-furan dicarboxylate) (PEF) show no persistent interactions, suggesting that these enzymes are not yet optimized for binding this alternative semiaromatic plastic polymer. Our study offers valuable insights into the structural stability of these enzymes and the molecular determinants driving PET binding onto their surfaces, sheds light on the mechanistic steps that precede the onset of hydrolysis, and provides a foundation for future enzyme optimization.
Collapse
Affiliation(s)
- Alessandro Berselli
- Department of Chemical and Geological Sciences (DSCG), University of Modena and Reggio Emilia (UNIMORE), Via Campi 103, 41125 Modena, Italy
| | - Maria Cristina Menziani
- Department of Chemical and Geological Sciences (DSCG), University of Modena and Reggio Emilia (UNIMORE), Via Campi 103, 41125 Modena, Italy
| | - Francesco Muniz-Miranda
- Department of Chemical and Geological Sciences (DSCG), University of Modena and Reggio Emilia (UNIMORE), Via Campi 103, 41125 Modena, Italy
| |
Collapse
|
40
|
dos Santos AM, da Costa CHS, Martins M, Goldbeck R, Skaf MS. Exploring the Structural and Dynamic Properties of a Chimeric Glycoside Hydrolase Protein in the Presence of Calcium Ions. Int J Mol Sci 2024; 25:11961. [PMID: 39596029 PMCID: PMC11594105 DOI: 10.3390/ijms252211961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
GH10 xylanases and GH62 Arabinofuranosidases play key roles in the breakdown of arabinoxylans and are important tools in various industrial and biotechnological processes, such as renewable biofuel production, the paper industry, and the production of short-chain xylooligosaccharides (XOS) from plant biomass. However, the use of these enzymes in industrial settings is often limited due to their relatively low thermostability and reduced catalytic efficiency. To overcome these limitations, strategies based on enzymatic chimera construction and the use of metal ions and other cofactors have been proposed to produce new recombinant enzymes with improved catalytic activity and thermostability. Here, we examine the conformational dynamics of a GH10-GH62 chimera at different calcium ion concentrations through molecular dynamics simulations. While experimental data have demonstrated improved activity and thermostability in GH10-GH62 chimera, the mechanistic basis for these enhancements remains unclear. We explored the structural details of the binding subsites of Ca2+ in the parental enzymes GH62 from Aspergillus fumigatus (Afafu62) and a recombinant GH10 from Cryptococcus flavescens (Xyn10cf), as well as their chimeric combination, and how negatively charged electron pairing located at the protein surface affects Ca2+ capture. The results indicate that Ca2+ binding significantly contributes to structural stability and catalytic cavity modulation in the chimera, particularly evident at a concentration of 0.01 M. This effect, not observed in the parental GH10 and GH62 enzymes, highlights how Ca2+ enhances stability in the overall chimeric enzyme, while supporting a larger cavity volume in the chimera GH62 subunit. The increased catalytic site volume and reduced structural flexibility in response to Ca2+ suggest that calcium binding minimizes non-productive conformational states, which could potentially improve catalytic turnover. The findings presented here may aid in the development of more thermostable and efficient catalytic systems.
Collapse
Affiliation(s)
- Alberto M. dos Santos
- Institute of Chemistry and Center for Computing in Engineering and Sciences, University of Campinas (UNICAMP), Campinas 13084-862, SP, Brazil; (A.M.d.S.); (C.H.S.d.C.)
| | - Clauber H. S. da Costa
- Institute of Chemistry and Center for Computing in Engineering and Sciences, University of Campinas (UNICAMP), Campinas 13084-862, SP, Brazil; (A.M.d.S.); (C.H.S.d.C.)
| | - Manoela Martins
- School of Food Engineering, University of Campinas (UNICAMP), Campinas 13083-862, SP, Brazil
| | - Rosana Goldbeck
- School of Food Engineering, University of Campinas (UNICAMP), Campinas 13083-862, SP, Brazil
| | - Munir S. Skaf
- Institute of Chemistry and Center for Computing in Engineering and Sciences, University of Campinas (UNICAMP), Campinas 13084-862, SP, Brazil; (A.M.d.S.); (C.H.S.d.C.)
| |
Collapse
|
41
|
Lai TT, Brooks CL. Accuracy and Reproducibility of Lipari-Szabo Order Parameters From Molecular Dynamics. J Phys Chem B 2024; 128:10813-10822. [PMID: 39466025 PMCID: PMC11790309 DOI: 10.1021/acs.jpcb.4c04895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The Lipari-Szabo generalized order parameter probes the picosecond to nanosecond time scale motions of a protein and is useful for rationalizing a multitude of biological processes such as protein recognition and ligand binding. Although these fast motions are an important and intrinsic property of proteins, it remains unclear what simulation conditions are most suitable to reproduce methyl symmetry axis side chain order parameter data (Saxis2) from molecular dynamics simulations. In this study, we show that, while Saxis2 tends to converge within tens of nanoseconds, it is essential to run 10 to 20 replicas starting from configurations close to the experimental structure to obtain the best agreement with experimental Saxis2 values. Additionally, in a comparison of force fields, AMBER ff14SB outperforms CHARMM36m in accurately capturing these fast time scale motions, and we suggest that the origin of this performance gap is likely attributed to differences in side chain torsional parametrization and not due to differences in the global protein conformations sampled by the force fields. This study provides insight into obtaining accurate and reproducible Saxis2 values from molecular simulations and underscores the necessity of using replica simulations to compute equilibrium properties.
Collapse
Affiliation(s)
- Thanh T. Lai
- Biophysics Program, University of Michigan, Ann Arbor, MI 48103
| | - Charles L. Brooks
- Department of Chemistry and Biophysics Program, University of Michigan, Ann Arbor, MI 48103
| |
Collapse
|
42
|
Dang T, EswarKumar N, Tripathi SK, Yan C, Wang CH, Cao M, Paul TK, Agboluaje EO, Xiong MP, Ivanov I, Ho MC, Zheng YG. Oligomerization of protein arginine methyltransferase 1 and its functional impact on substrate arginine methylation. J Biol Chem 2024; 300:107947. [PMID: 39491649 DOI: 10.1016/j.jbc.2024.107947] [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: 06/09/2024] [Revised: 10/20/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
Protein arginine methyltransferases (PRMTs) are important posttranslational modifying enzymes in eukaryotic proteins and regulate diverse pathways from gene transcription, RNA splicing, and signal transduction to metabolism. Increasing evidence supports that PRMTs exhibit the capacity to form higher-order oligomeric structures, but the structural basis of PRMT oligomerization and its functional consequence are elusive. Herein, we revealed for the first time different oligomeric structural forms of the predominant arginine methyltransferase PRMT1 using cryo-EM, which included tetramer (dimer of dimers), hexamer (trimer of dimers), octamer (tetramer of dimers), decamer (pentamer of dimers), and also helical filaments. Through a host of biochemical assays, we showed that PRMT1 methyltransferase activity was substantially enhanced as a result of the high-ordered oligomerization. High-ordered oligomerization increased the catalytic turnover and the multimethylation processivity of PRMT1. Presence of a catalytically dead PRMT1 mutant also enhanced the activity of WT PRMT1, pointing out a noncatalytic role of oligomerization. Structural modeling demonstrates that oligomerization enhances substrate retention at the PRMT1 surface through electrostatic force. Our studies offered key insights into PRMT1 oligomerization and established that oligomerization constitutes a novel molecular mechanism that positively regulates the enzymatic activity of PRMTs in biology.
Collapse
Affiliation(s)
- Tran Dang
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia, United States
| | | | - Sunil Kumar Tripathi
- Department of Chemistry, Georgia State University, Atlanta, Georgia, USA; Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Chunli Yan
- Department of Chemistry, Georgia State University, Atlanta, Georgia, USA; Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Chun-Hsiung Wang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Mengtong Cao
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia, United States
| | - Tanmoy Kumar Paul
- Department of Chemistry, Georgia State University, Atlanta, Georgia, USA; Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Elizabeth Oladoyin Agboluaje
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia, United States
| | - May P Xiong
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia, United States
| | - Ivaylo Ivanov
- Department of Chemistry, Georgia State University, Atlanta, Georgia, USA; Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Meng-Chiao Ho
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan; Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan; Graduate Institute of Biochemistry and Molecular Biology, National Taiwan University, Taipei, Taiwan.
| | - Y George Zheng
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, Georgia, United States.
| |
Collapse
|
43
|
Roth O, Yechezkel S, Serero O, Eliyahu A, Vints I, Tzeela P, Carignano A, Janacek DP, Peters V, Kessel A, Dwivedi V, Carmeli-Weissberg M, Shaya F, Faigenboim-Doron A, Ung KL, Pedersen BP, Riov J, Klavins E, Dawid C, Hammes UZ, Ben-Tal N, Napier R, Sadot E, Weinstain R. Slow release of a synthetic auxin induces formation of adventitious roots in recalcitrant woody plants. Nat Biotechnol 2024; 42:1705-1716. [PMID: 38267759 DOI: 10.1038/s41587-023-02065-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 11/15/2023] [Indexed: 01/26/2024]
Abstract
Clonal propagation of plants by induction of adventitious roots (ARs) from stem cuttings is a requisite step in breeding programs. A major barrier exists for propagating valuable plants that naturally have low capacity to form ARs. Due to the central role of auxin in organogenesis, indole-3-butyric acid is often used as part of commercial rooting mixtures, yet many recalcitrant plants do not form ARs in response to this treatment. Here we describe the synthesis and screening of a focused library of synthetic auxin conjugates in Eucalyptus grandis cuttings and identify 4-chlorophenoxyacetic acid-L-tryptophan-OMe as a competent enhancer of adventitious rooting in a number of recalcitrant woody plants, including apple and argan. Comprehensive metabolic and functional analyses reveal that this activity is engendered by prolonged auxin signaling due to initial fast uptake and slow release and clearance of the free auxin 4-chlorophenoxyacetic acid. This work highlights the utility of a slow-release strategy for bioactive compounds for more effective plant growth regulation.
Collapse
Affiliation(s)
- Ohad Roth
- School of Plant Sciences and Food Security, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Sela Yechezkel
- The Institute of Plant Sciences, The Volcani Center, Ministry of Agriculture and Rural Development, Rishon LeZion, Israel
| | - Ori Serero
- The Institute of Plant Sciences, The Volcani Center, Ministry of Agriculture and Rural Development, Rishon LeZion, Israel
- The Robert H. Smith Institute of Plant Sciences and Genetics in Agriculture, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Avi Eliyahu
- The Institute of Plant Sciences, The Volcani Center, Ministry of Agriculture and Rural Development, Rishon LeZion, Israel
- The Robert H. Smith Institute of Plant Sciences and Genetics in Agriculture, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Inna Vints
- School of Plant Sciences and Food Security, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Pan Tzeela
- The Institute of Plant Sciences, The Volcani Center, Ministry of Agriculture and Rural Development, Rishon LeZion, Israel
- The Robert H. Smith Institute of Plant Sciences and Genetics in Agriculture, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Alberto Carignano
- Department of Electrical and Computer Engineering, University of Washington, Seattle, WA, USA
| | - Dorina P Janacek
- Chair of Plant Systems Biology, Technical University of Munich, Freising, Germany
| | - Verena Peters
- Chair of Food Chemistry and Molecular and Sensory Science, Technical University of Munich, Freising, Germany
| | - Amit Kessel
- Department of Biochemistry and Molecular BiologySchool of Neurobiology, Biochemistry & Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Vikas Dwivedi
- The Institute of Plant Sciences, The Volcani Center, Ministry of Agriculture and Rural Development, Rishon LeZion, Israel
| | - Mira Carmeli-Weissberg
- The Institute of Plant Sciences, The Volcani Center, Ministry of Agriculture and Rural Development, Rishon LeZion, Israel
| | - Felix Shaya
- The Institute of Plant Sciences, The Volcani Center, Ministry of Agriculture and Rural Development, Rishon LeZion, Israel
| | - Adi Faigenboim-Doron
- The Institute of Plant Sciences, The Volcani Center, Ministry of Agriculture and Rural Development, Rishon LeZion, Israel
| | - Kien Lam Ung
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | - Joseph Riov
- The Robert H. Smith Institute of Plant Sciences and Genetics in Agriculture, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Eric Klavins
- Department of Electrical and Computer Engineering, University of Washington, Seattle, WA, USA
| | - Corinna Dawid
- Chair of Food Chemistry and Molecular and Sensory Science, Technical University of Munich, Freising, Germany
| | - Ulrich Z Hammes
- Chair of Plant Systems Biology, Technical University of Munich, Freising, Germany
| | - Nir Ben-Tal
- Department of Biochemistry and Molecular BiologySchool of Neurobiology, Biochemistry & Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Richard Napier
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Einat Sadot
- The Institute of Plant Sciences, The Volcani Center, Ministry of Agriculture and Rural Development, Rishon LeZion, Israel.
| | - Roy Weinstain
- School of Plant Sciences and Food Security, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
44
|
Staar M, Ahlborn L, Estévez-Gay M, Pallasch K, Osuna S, Schallmey A. A Dynamic Loop in Halohydrin Dehalogenase HheG Regulates Activity and Enantioselectivity in Epoxide Ring Opening. ACS Catal 2024; 14:15976-15987. [PMID: 39507489 PMCID: PMC11536340 DOI: 10.1021/acscatal.4c04815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/30/2024] [Accepted: 10/03/2024] [Indexed: 11/08/2024]
Abstract
Halohydrin dehalogenase HheG and its homologues are remarkable enzymes for the efficient ring opening of sterically demanding internal epoxides using a variety of nucleophiles. The enantioselectivity of the respective wild-type enzymes, however, is usually insufficient for application and frequently requires improvement by protein engineering. We herein demonstrate that the highly flexible N-terminal loop of HheG, comprising residues 39 to 47, has a tremendous impact on the activity as well as enantioselectivity of this enzyme in the ring opening of structurally diverse epoxide substrates. Thus, highly active and enantioselective HheG variants could be accessed through targeted engineering of this loop. In this regard, variant M45F displayed almost 10-fold higher specific activity than wild type in the azidolysis of cyclohexene oxide, yielding the corresponding product (1S,2S)-2-azidocyclohexan-1-ol in 96%eeP (in comparison to 49%eeP for HheG wild type). Moreover, this variant was also improved regarding activity and enantioselectivity in the ring opening of cyclohexene oxide with other nucleophiles, demonstrating even inverted enantioselectivity with cyanide and cyanate. In contrast, a complete loop deletion yielded an inactive enzyme. Concomitant computational analyses of HheG M45F in comparison to wild type enzyme revealed that mutation M45F promotes the productive binding of cyclohexene oxide and azide in the active site by establishing noncovalent C-H ··π interactions between epoxide and F45. These interactions further position one of the two carbon atoms of the epoxide ring closer to the azide, resulting in higher enantioselectivity. Additionally, stable and enantioselective cross-linked enzyme crystals of HheG M45F were successfully generated after combination with mutation D114C. Overall, our study highlights that a highly flexible loop in HheG governs the enzyme's activity and selectivity in epoxide ring opening and should thus be considered in future protein engineering campaigns of HheG.
Collapse
Affiliation(s)
- Marcel Staar
- Institute
for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany
| | - Lina Ahlborn
- Institute
for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany
| | - Miquel Estévez-Gay
- CompBioLab
Group, Institut de Química Computacional i Catàlisi
(IQCC), Departament de Química, Universitat
de Girona, c/Maria Aurèlia Capmany 69, 17003 Girona, Catalonia, Spain
| | - Katharina Pallasch
- Institute
for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany
| | - Sílvia Osuna
- CompBioLab
Group, Institut de Química Computacional i Catàlisi
(IQCC), Departament de Química, Universitat
de Girona, c/Maria Aurèlia Capmany 69, 17003 Girona, Catalonia, Spain
- ICREA, Passeig Lluís Companys 23, 08010 Barcelona, Catalonia, Spain
| | - Anett Schallmey
- Institute
for Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany
- Zentrum
für Pharmaverfahrenstechnik (PVZ), Technische Universität Braunschweig, Franz-Liszt-Str. 35a, 38106 Braunschweig, Germany
- Braunschweig
Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
| |
Collapse
|
45
|
Alexopoulos S, McGawley M, Mathews R, Papakostopoulou S, Koulas S, Leonidas DD, Zwain T, Hayes JM, Skamnaki V. Evidence for the Quercetin Binding Site of Glycogen Phosphorylase as a Target for Liver-Isoform-Selective Inhibitors against Glioblastoma: Investigation of Flavanols Epigallocatechin Gallate and Epigallocatechin. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:24070-24081. [PMID: 39433280 PMCID: PMC11528470 DOI: 10.1021/acs.jafc.4c06920] [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: 08/01/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024]
Abstract
Glycogen phosphorylase (GP) is the rate-determining enzyme in glycogenolysis, and its druggability has been extensively studied over the years for the development of therapeutics against type 2 diabetes (T2D) and, more recently, cancer. However, the conservation of binding sites between the liver and muscle isoforms makes the inhibitor selectivity challenging. Using a combination of kinetic, crystallographic, modeling, and cellular studies, we have probed the binding of dietary flavonoids epigallocatechin gallate (EGCG) and epigallocatechin (EGC) to GP isoforms. The structures of rmGPb-EGCG and rmGPb-EGC complexes were determined by X-ray crystallography, showing binding at the quercetin binding site (QBS) in agreement with kinetic studies that revealed both compounds as noncompetitive inhibitors of GP, with EGCG also causing a significant reduction in cell viability and migration of U87-MG glioblastoma cells. Interestingly, EGCG exhibits different binding modes to GP isoforms, revealing QBS as a promising site for GP targeting, offering new opportunities for the design of liver-selective GP inhibitors.
Collapse
Affiliation(s)
- Serafeim Alexopoulos
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| | - Megan McGawley
- School
of Pharmacy & Biomedical Sciences, University
of Central Lancashire, Preston PR1 2HE, U.K.
| | - Roshini Mathews
- School
of Pharmacy & Biomedical Sciences, University
of Central Lancashire, Preston PR1 2HE, U.K.
| | - Souzana Papakostopoulou
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| | - Symeon Koulas
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| | - Demetres D. Leonidas
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| | - Tamara Zwain
- School
of Pharmacy & Biomedical Sciences, University
of Central Lancashire, Preston PR1 2HE, U.K.
| | - Joseph M. Hayes
- School
of Pharmacy & Biomedical Sciences, University
of Central Lancashire, Preston PR1 2HE, U.K.
| | - Vasiliki Skamnaki
- Department
of Biochemistry and Biotechnology, University
of Thessaly, Biopolis, Larisa 41500, Greece
| |
Collapse
|
46
|
Weimar J, Hirschmann F, Oettel M. Effective patchiness from critical points of a coarse-grained protein model with explicit shape and charge anisotropy. SOFT MATTER 2024; 20:8455-8467. [PMID: 39400290 DOI: 10.1039/d4sm00867g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Colloidal model systems are successful in rationalizing emergent phenomena like aggregation, rheology and phase behaviour of protein solutions. Colloidal theory in conjunction with isotropic interaction models is often employed to estimate the stability of such solutions. In particular, a universal criterion for the reduced second virial coefficient at the critical point is frequently invoked which is based on the behavior of short-range attractive fluids (Noro-Frenkel rule, ). However, if anisotropic models for the protein-protein interaction are considered, e.g. the Kern-Frenkel (KF) patchy particle model, the value of the criterion is shifted to lower values and explicitly depends on the number of patches. If an explicit shape anisotropy is considered, as e.g. in a coarse-grained protein model, the normalization of becomes ambiguous to some extent, as no unique exclusion volume can be defined anymore. Here, we investigate a low-resolution, coarse-grained model for the globular protein bovine serum albumin (BSA) and study effects of charge-anisotropy on the phase diagram (determined by simulations) at the isoelectric point. We present methods of assigning an "effective patchiness" to our protein model by comparing its critical properties to the KF model. We find that doubling the native charges increases the critical temperature Tc by ≈14% and that our BSA model can be compared to a 3 to 5 patch KF model. Finally, we argue that applying existing criteria from colloidal theory should be done with care, due to multiple, physically plausible ways of how to assign effective diameters to shape-anisotropic models.
Collapse
Affiliation(s)
- Jens Weimar
- Institute for Applied Physics, University of Tübingen, Auf der Morgenstelle 10, 72076 Tübingen, Germany.
| | - Frank Hirschmann
- Institute for Applied Physics, University of Tübingen, Auf der Morgenstelle 10, 72076 Tübingen, Germany.
| | - Martin Oettel
- Institute for Applied Physics, University of Tübingen, Auf der Morgenstelle 10, 72076 Tübingen, Germany.
| |
Collapse
|
47
|
Stokely AM, Votapka LW, Hock MT, Teitgen AE, McCammon JA, McCulloch AD, Amaro RE. NetSci: A Library for High Performance Biomolecular Simulation Network Analysis Computation. J Chem Inf Model 2024; 64:7966-7976. [PMID: 39364881 DOI: 10.1021/acs.jcim.4c00899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
We present the NetSci program-an open-source scientific software package designed for estimating mutual information (MI) between data sets using GPU acceleration and a k-nearest-neighbor algorithm. This approach significantly enhances calculation speed, achieving improvements of several orders of magnitude over traditional CPU-based methods, with data set size limits dictated only by available hardware. To validate NetSci, we accurately compute MI for an analytically verifiable two-dimensional Gaussian distribution and replicate the generalized correlation (GC) analysis previously conducted on the B1 domain of protein G. We also apply NetSci to molecular dynamics simulations of the Sarcoendoplasmic Reticulum Calcium-ATPase (SERCA) pump, exploring the allosteric mechanisms and pathways influenced by ATP and 2'-deoxy-ATP (dATP) binding. Our analysis reveals distinct allosteric effects induced by ATP compared to dATP, with predicted information pathways from the bound nucleotide to the calcium-binding domain differing based on the nucleotide involved. NetSci proves to be a valuable tool for estimating MI and GC in various data sets and is particularly effective for analyzing intraprotein communication and information transfer.
Collapse
Affiliation(s)
- Andrew M Stokely
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, United States
- Mesoscale and Microscale Meteorology Laboratory, National Center for Atmospheric Research, Boulder, Colorado 80307, United States
| | - Lane W Votapka
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, United States
| | - Marcus T Hock
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Abigail E Teitgen
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
| | - J Andrew McCammon
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, California 92093, United States
- Department of Pharmacology, University of California San Diego, La Jolla, California 92093, United States
| | - Andrew D McCulloch
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Rommie E Amaro
- Department of Molecular Biology, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
48
|
Chen J, Qi S, Wang Z, Hu L, Liu J, Huang G, Peng Y, Fang Z, Wu Q, Hu Y, Guo K. Ene-Reductase-Catalyzed Aromatization of Simple Cyclohexanones to Phenols. Angew Chem Int Ed Engl 2024; 63:e202408359. [PMID: 39106109 DOI: 10.1002/anie.202408359] [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: 05/17/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 08/09/2024]
Abstract
Direct aromatization of cyclohexanones to synthesize substituted phenols represents a significant challenge in modern synthetic chemistry. Herein, we describe a novel ene-reductase (TsER) catalytic system that converts substituted cyclohexanones into the corresponding phenols. This process involves the successive dehydrogenation of two saturated carbon-carbon bonds within the six-membered ring of cyclohexanones and utilizes molecular oxygen to drive the reaction cycle. It demonstrates a versatile and efficient approach for the synthesis of substituted phenols, providing a valuable complement to existing chemical methodologies.
Collapse
Affiliation(s)
- Jie Chen
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 211816, PR China
| | - Shaofang Qi
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 211816, PR China
| | - Zhiguo Wang
- Institute of Aging Research, Hangzhou Normal University, Zhejiang, Hangzhou, 311121, PR China
| | - Liran Hu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 211816, PR China
| | - Jialing Liu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 211816, PR China
| | - Guixiang Huang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 211816, PR China
| | - Yongzhen Peng
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 211816, PR China
| | - Zheng Fang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 211816, PR China
| | - Qi Wu
- Department of Chemistry, Zhejiang University, Zhejiang, Hangzhou, 310027, PR China
| | - Yujing Hu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 211816, PR China
| | - Kai Guo
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 211816, PR China
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 210009, PR China
| |
Collapse
|
49
|
Ferrer S, Moliner V, Świderek K. Electrostatic Preorganization in Three Distinct Heterogeneous Proteasome β-Subunits. ACS Catal 2024; 14:15237-15249. [PMID: 39444531 PMCID: PMC11494509 DOI: 10.1021/acscatal.4c04964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/08/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024]
Abstract
The origin of the enzyme's powerful role in accelerating chemical reactions is one of the most critical and still widely discussed questions. It is already accepted that enzymes impose an electrostatic field onto their substrates by adopting complex three-dimensional structures; therefore, the preorganization of electric fields inside protein active sites has been proposed as a crucial contributor to catalytic mechanisms and rate constant enhancement. In this work, we focus on three catalytically active β-subunits of 20S proteasomes with low sequence identity (∼30%) whose active sites, although situated in an electrostatically miscellaneous environment, catalyze the same chemical reaction with similar catalytic efficiency. Our in silico experiments reproduce the experimentally observed equivalent reactivity of the three sites and show that obliteration of the electrostatic potential in all active sites would deprive the enzymes of their catalytic power by slowing down the chemical process by a factor of 1035. To regain enzymatic efficiency, besides catalytic Thr1 and Lys33 residues, the presence of aspartic acid in position 17 and an aqueous solvent is required, proving that the electrostatic potential generated by the remaining residues is insignificant for catalysis. Moreover, it was found that the gradual decay of atomic charges on Asp17 strongly correlates with the enzyme's catalytic rate deterioration as well as with a change in the charge distributions due to introduced mutations. The computational procedure used and described here may help identify key residues for catalysis in other biomolecular systems and consequently may contribute to the process of designing enzyme-like synthetic catalysts.
Collapse
Affiliation(s)
- Silvia Ferrer
- BioComp Group, Institute
of Advanced Materials (INAM), Universitat
Jaume I, Avenida de Vicent Sos Baynat, s/n, 12071 Castellón, Spain
| | - Vicent Moliner
- BioComp Group, Institute
of Advanced Materials (INAM), Universitat
Jaume I, Avenida de Vicent Sos Baynat, s/n, 12071 Castellón, Spain
| | - Katarzyna Świderek
- BioComp Group, Institute
of Advanced Materials (INAM), Universitat
Jaume I, Avenida de Vicent Sos Baynat, s/n, 12071 Castellón, Spain
| |
Collapse
|
50
|
Hu Y, Li G, Zhao S, Dai J, Peng X, Zhao Q. Charge transfer characteristics in rhodopsin mimics during photoexcitation. Phys Chem Chem Phys 2024; 26:26004-26011. [PMID: 39370953 DOI: 10.1039/d4cp02970d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
To gain insights into the light-harvesting capabilities of the chromophores, it is essential to understand their molecular and electronic structures within their natural chemical or biological contexts. Rhodopsins display varied absorption characteristics due to the interaction between the chromophore retinal and its surrounding protein environments. In this study, we employed a quantum mechanics/molecular mechanics approach to examine a series of artificially designed rhodopsin mimics based on human cellular retinol acid binding protein 2 (hCRABP II). We elucidated the electron transfer within the all-trans protonated Schiff base upon light excitation, and our calculated absorption spectra show well consistency with the experimental result. Furthermore, the interaction mechanisms between the chromophore and the protein were investigated, and the relationship between the blueshifts and redshifts in the absorption spectra was analyzed. Our calculation demonstrates that the blueshifts and redshifts in the rhodopsin mimics correlate well with attractive (such as the hydrogen bonds or electrostatic interactions) and repulsive interactions (such as the steric effects) between the chromophore and the protein environment, respectively. These findings could provide hints for designing rhodopsin with absorption spectra at different wavelengths.
Collapse
Affiliation(s)
- Yongnan Hu
- Center for Quantum Technology Research, Key Laboratory of Advanced Optoelectronic Quantum Architecture and Measurements (MOE), School of Physics, Beijing Institute of Technology, Beijing 100081, China
| | - Gaoshang Li
- Center for Quantum Technology Research, Key Laboratory of Advanced Optoelectronic Quantum Architecture and Measurements (MOE), School of Physics, Beijing Institute of Technology, Beijing 100081, China
| | - Siteng Zhao
- Center for Quantum Technology Research, Key Laboratory of Advanced Optoelectronic Quantum Architecture and Measurements (MOE), School of Physics, Beijing Institute of Technology, Beijing 100081, China
| | - Jin Dai
- Center for Quantum Technology Research, Key Laboratory of Advanced Optoelectronic Quantum Architecture and Measurements (MOE), School of Physics, Beijing Institute of Technology, Beijing 100081, China
| | - Xubiao Peng
- Center for Quantum Technology Research, Key Laboratory of Advanced Optoelectronic Quantum Architecture and Measurements (MOE), School of Physics, Beijing Institute of Technology, Beijing 100081, China
- Beijing Academy of Quantum Information Sciences, Beijing 100193, China.
| | - Qing Zhao
- Center for Quantum Technology Research, Key Laboratory of Advanced Optoelectronic Quantum Architecture and Measurements (MOE), School of Physics, Beijing Institute of Technology, Beijing 100081, China
- Beijing Academy of Quantum Information Sciences, Beijing 100193, China.
| |
Collapse
|