1
|
Cao S, Nüske F, Liu B, Soley MB, Huang X. AMUSET-TICA: A Tensor-Based Approach for Identifying Slow Collective Variables in Biomolecular Dynamics. J Chem Theory Comput 2025; 21:4855-4866. [PMID: 40254940 DOI: 10.1021/acs.jctc.5c00076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Elucidating collective variables (CVs) for biomolecular dynamics is crucial for understanding numerous biological processes. By leveraging the tensor-train data structure, a multilinear version of the AMUSE (Algorithm for Multiple Unknown Signals) algorithm for Koopman approximation (AMUSEt) was recently developed to identify CVs for biomolecular dynamics. To find slow CVs, AMUSEt transforms input features (e.g., pairwise atomic distances) into nonlinear basis functions (e.g., Gaussian functions) and encodes these nonlinear basis functions within a tensor-train structure via time-lagged correlation functions. Due to the need to fit these tensor-train data structures into computer memory, AMUSEt can handle only a limited number of input features. Consequently, AMUSEt relies on manually selecting and ranking features based on physical intuition to fully capture the slow dynamics. However, when applied to complex biological systems with numerous features, this selection and ranking process becomes increasingly challenging. To address this challenge, here we present AMUSET-TICA (AMUSEt-based Time-lagged Independent Component Analysis), a CV-identification method using time-structure-independent components (tICs) as the input features for AMUSEt. The key insight of AMUSET-TICA lies in its highly effective embedding of high-dimensional atomistic protein conformations, achieved by expanding orthogonal tICs into overlapping Gaussian basis functions through a tensor-product data structure. This eliminates the need for manually selecting and ranking input features for a wide range of biomolecular systems. We demonstrate that AMUSET-TICA consistently and significantly outperforms AMUSEt and tICA in identifying slow CVs for three different biomolecular systems: alanine dipeptide, the N-terminal domain of L9 (NTL9), and the FIP35 WW domain. For all these systems, the CVs generated by AMUSET-TICA accurately describe the slowest dynamical modes underlying these biological conformational changes. Furthermore, we show that AMUSET-TICA achieves performance comparable to deep-learning approaches like VAMPnets in identifying the slowest dynamical modes, while being significantly more computationally efficient in terms of CPU time. In addition, the CVs yielded by AMUSET-TICA provide insights into the folding mechanisms of NTL9 and the FIP35 WW domain, including CV3 and CV4 of the WW domain, which capture its two parallel folding pathways. We expect AMUSET-TICA can be widely applied to facilitate the investigation of biomolecular dynamics.
Collapse
Affiliation(s)
- Siqin Cao
- Department of Chemistry, Theoretical Chemistry Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Feliks Nüske
- Max-Planck-Institute for Dynamics of Complex Technical Systems, Magdeburg 39106, Germany
| | - Bojun Liu
- Department of Chemistry, Theoretical Chemistry Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Micheline B Soley
- Department of Chemistry, Theoretical Chemistry Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| | - Xuhui Huang
- Department of Chemistry, Theoretical Chemistry Institute, University of Wisconsin-Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
2
|
Sundaresan S, Raghuvamsi PV, Rathinavelan T. MDTAP: a tool to analyze permeation events across membrane proteins. BIOINFORMATICS ADVANCES 2025; 5:vbaf102. [PMID: 40421423 PMCID: PMC12104522 DOI: 10.1093/bioadv/vbaf102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/08/2025] [Indexed: 05/28/2025]
Abstract
Motivation Molecular dynamics (MD) simulations provide critical insights into the transport of solutes, solvents, and drug molecules across protein channels embedded in a membrane bilayer. However, identifying and analyzing the permeation events from complex simulation data remains as a challenging and laborious task. Thus, an automated tool that facilitates the capture of permeation events of any molecular type across any channel is essential to streamline MD trajectory analysis and enhance the understanding of biological processes in a timely manner. Results Molecular Dynamics Trajectory Analysis of Permeation (MDTAP) is a Linux/Mac-based software that automatically detects permeation events across membrane-embedded protein and nucleic acid channels. The tool accepts trajectories in DCD (CHARMM/NAMD) and PDB format (obtained from any MD simulation package) and employs bash scripts to analyze the input trajectories to characterize the molecular permeation. The efficiency of MDTAP is demonstrated using MD trajectories of Escherichia coli outer membrane protein Wzi and E. coli Aquaporin Z. MDTAP can also analyze permeation across heterogeneous lipid membranes and artificial nucleic acid channels, addressing their growing importance. Thus, MDTAP simplifies trajectory analysis and also reduces the need for manual inspection. Availability and implementation MDTAP is open-source and is freely available on GitHub (https://github.com/MBL-lab/MDTAP), including source code, installation instructions, and usage documentation.
Collapse
Affiliation(s)
- Sruthi Sundaresan
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | - Palur Venkata Raghuvamsi
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi, Telangana 502284, India
| | | |
Collapse
|
3
|
Tang CL, Li YQ, Du XK, Fang XX, Guang YM, Li PZ, Chen S, Xue SY, Yu JM, Liu XY, Luo YP, Zhou LX, Luo C, Xiong H, Liang ZJ, Ding H. Identifying a non-conserved site for achieving allosteric covalent inhibition of CECR2. Acta Pharmacol Sin 2025; 46:1476-1491. [PMID: 39833305 PMCID: PMC12032100 DOI: 10.1038/s41401-024-01452-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025]
Abstract
The bromodomain (BRD) represents a highly conserved structural module that provides BRD proteins with fundamental functionality in modulating protein-protein interactions involved in diverse biological processes such as chromatin-mediated gene transcription, DNA recombination, replication and repair. Consequently, dysregulation of BRD proteins has been implicated in the pathogenesis of numerous human diseases. In recent years, considerable scientific endeavors have focused on unraveling the molecular mechanisms underlying BRDs and developing inhibitors that target these domains. While these inhibitors compete for binding with the acetylated lysine binding site of BRDs, achieving inhibition of BRD proteins via competitive pocket binding has proven challenging due to the conserved nature of these pockets. To address this limitation, the present study employed dynamic simulations for a comprehensive analysis, leading to the identification of a non-conserved pocket in CECR2 for achieving BRD family inhibition through allosteric modulation. Subsequently, the compound BAY 11-7085 was proven capable of covalently binding to C494 of this pocket after covalent docking and biological verification in vitro. The allosteric inhibition strategy of CECR2 was further verified by the structurally optimized compound LC-CE-7, which is an allosteric covalent CECR2 inhibitor with anti-cancer effects in MDA-MB-231 cells.
Collapse
Affiliation(s)
- Cai-Ling Tang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yuan-Qing Li
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xi-Kun Du
- Center for Systems Biology, Department of Bioinformatics, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Xiao-Xia Fang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Yi-Man Guang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Pei-Zhuo Li
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Shuang Chen
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Sheng-Yu Xue
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jia-Min Yu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiao-Yi Liu
- Center for Systems Biology, Department of Bioinformatics, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China
| | - Yi-Pan Luo
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
- School of Pharmacy, Zunyi Medical University, Zunyi, 563000, China
| | - Lan-Xin Zhou
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
- School of Pharmacy, Guizhou Medical University, Guiyang, 550004, China
| | - Cheng Luo
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
- School of Pharmacy, Guizhou Medical University, Guiyang, 550004, China
| | - Huan Xiong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
| | - Zhong-Jie Liang
- Center for Systems Biology, Department of Bioinformatics, School of Life Sciences, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, 215123, China.
| | - Hong Ding
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmacy, Guizhou Medical University, Guiyang, 550004, China.
| |
Collapse
|
4
|
Vornweg JR, Maier TM, Jacob CR. The density-based many-body expansion for poly-peptides and proteins. Phys Chem Chem Phys 2025; 27:8719-8730. [PMID: 40235457 DOI: 10.1039/d5cp00727e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Fragmentation schemes enable the efficient quantum-chemical treatment of large biomolecular systems, and provide an ideal starting point for the development of accurate machine-learning potentials for proteins. Here, we present a fragment-based method that only uses calculations for single-amino acids and their dimers, and is able to reduce the fragmentation error in total energies to ca. 1 kJ mol-1 per amino acid for polypeptides and proteins across different structural motifs. This is achieved by combining a two-body extension of the molecular fractionation with conjugate caps (MFCC) scheme with the density-based many-body expansion (db-MBE), thus extending the applicability of the db-MBE from molecular clusters to polypeptides and proteins.
Collapse
Affiliation(s)
- Johannes R Vornweg
- Technische Universität Braunschweig, Institute of Physical and Theoretical Chemistry, Gaußstraße 17, 38106 Braunschweig, Germany.
| | - Toni M Maier
- Technische Universität Braunschweig, Institute of Physical and Theoretical Chemistry, Gaußstraße 17, 38106 Braunschweig, Germany.
| | - Christoph R Jacob
- Technische Universität Braunschweig, Institute of Physical and Theoretical Chemistry, Gaußstraße 17, 38106 Braunschweig, Germany.
| |
Collapse
|
5
|
Mbah Bake M, Tsahnang Fofack HM, Nouping Fekoua JN, Ntie-Kang F, Pierre Vercauteren D, Mbouombouo Ndassa I, Mohammad-Salim HA, de Julián-Ortiz JV. Identification of Potential Inhibitors of Plasmodium falciparum L-lactate Dehydrogenase From Selected African Compound Libraries: Virtual Screening, Molecular Mechanics-Generalized Born Surface Area, and Molecular Dynamics Studies. Chem Biodivers 2025:e01004. [PMID: 40289738 DOI: 10.1002/cbdv.202501004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 04/28/2025] [Indexed: 04/30/2025]
Abstract
We evaluated 4,512 natural products from natural product library from Central African medicinal plants for drug discovery and South African natural compounds database libraries for the identification of potential Plasmodium falciparum L-lactate dehydrogenase inhibitors considering the virtual screening process. Extra precision virtual screening enabled the ranking of the top hundred hit molecules based on their docking properties. The selected hits were further shortened based on docking and molecular mechanics-generalized born surface area parameters in comparison with the reference. As a result, four hits were chosen: Mol1, Mol2, Mol3, and Mol4, all of them from the chalcone and quinone families. These molecules showed good predicted absorption, distribution, metabolism, and excretion, and toxicity properties. Finally, the molecular dynamics simulation results showed that the three chalcones, Mol1-4, formed an H-bond, hydrophobic interaction with key amino acids in the active site. This in silico study suggests that the chalcone compounds could serve as a potential source for developing new effective antimalarial drugs to combat malaria. Further in vitro or in vivo studies might be conducted to determine their actual effectiveness.
Collapse
Affiliation(s)
- Maraf Mbah Bake
- Faculty of Science, Physical and Theoretical Chemistry Unit, Laboratory of Applied Physical and Analytical Chemistry, University of Yaoundé I, Yaounde, Cameroon
- Department of Chemistry, Computational Chemistry Laboratory, Higher Teacher Training College, University of Yaoundé I, Yaounde, Cameroon
| | - Hans Merlin Tsahnang Fofack
- Faculté des Sciences, Laboratoire Optique et Applications, Centre de Physique Atomique Moléculaire et Optique Quantique, Université de Douala, Douala, Cameroon
- Faculty of Science, Department of Chemistry, Analytical, Structural and Materials Chemistry Laboratory, University of Douala, Douala, Cameroon
| | - Joëlle Nadia Nouping Fekoua
- Faculty of Science, Physical and Theoretical Chemistry Unit, Laboratory of Applied Physical and Analytical Chemistry, University of Yaoundé I, Yaounde, Cameroon
- Department of Chemistry, Computational Chemistry Laboratory, Higher Teacher Training College, University of Yaoundé I, Yaounde, Cameroon
| | - Fidele Ntie-Kang
- Center for Drug Discovery & Department of Chemistry, Faculty of Science, University of Buea, Buea, Cameroon
| | - Daniel Pierre Vercauteren
- Département de chimie, Namur Institute of Structured Matter (NISM), Namur Research Institute for Life Sciences (Narilis), NAmur MEdicine & Drug Innovation Center (NAMEDIC), Université de Namur, Namur, Belgium
| | - Ibrahim Mbouombouo Ndassa
- Department of Chemistry, Computational Chemistry Laboratory, Higher Teacher Training College, University of Yaoundé I, Yaounde, Cameroon
- Faculty of Science, Department of Applied Chemistry, University of Ebolowa, Ebolowa, Cameroon
| | - Haydar A Mohammad-Salim
- TCCG Lab, Scientific Research Center, University of Zakho, Zakho, Iraq
- Department of Chemistry, College of Science, University of Zakho, Zakho, Iraq
- Department of Physical Chemistry, Pharmacy Faculty, University of Valencia, Valencia, Spain
| | | |
Collapse
|
6
|
Pant S, Dehghani-Ghahnaviyeh S, Trebesch N, Rasouli A, Chen T, Kapoor K, Wen PC, Tajkhorshid E. Dissecting Large-Scale Structural Transitions in Membrane Transporters Using Advanced Simulation Technologies. J Phys Chem B 2025; 129:3703-3719. [PMID: 40100959 DOI: 10.1021/acs.jpcb.5c00104] [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: 03/20/2025]
Abstract
Membrane transporters are integral membrane proteins that act as gatekeepers of the cell, controlling fundamental processes such as recruitment of nutrients and expulsion of waste material. At a basic level, transporters operate using the "alternating access model," in which transported substances are accessible from only one side of the membrane at a time. This model usually involves large-scale structural changes in the transporter, which often cannot be captured using unbiased, conventional molecular simulation techniques. In this article, we provide an overview of some of the major simulation techniques that have been applied to characterize the structural dynamics and energetics involved in the transition of membrane transporters between their functional states. After briefly introducing each technique, we discuss some of their advantages and limitations and provide some recent examples of their application to membrane transporters.
Collapse
Affiliation(s)
- Shashank Pant
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801-3028, United States
| | - Sepehr Dehghani-Ghahnaviyeh
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801-3028, United States
| | - Noah Trebesch
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801-3028, United States
| | - Ali Rasouli
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801-3028, United States
| | - Tianle Chen
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801-3028, United States
| | - Karan Kapoor
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801-3028, United States
| | - Po-Chao Wen
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801-3028, United States
| | - Emad Tajkhorshid
- Theoretical and Computational Biophysics Group, NIH Resource for Macromolecular Modeling and Visualization, Beckman Institute for Advanced Science and Technology, Department of Biochemistry, and Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois 61801-3028, United States
| |
Collapse
|
7
|
Gheeraert A, Leroux V, Mias-Lucquin D, Karami Y, Vuillon L, Chauvot de Beauchêne I, Devignes MD, Rivalta I, Maigret B, Chaloin L. Subtle Changes at the RBD/hACE2 Interface During SARS-CoV-2 Variant Evolution: A Molecular Dynamics Study. Biomolecules 2025; 15:541. [PMID: 40305276 PMCID: PMC12024731 DOI: 10.3390/biom15040541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
The SARS-CoV-2 Omicron variants show different behavior compared to the previous variants, especially with respect to the Delta variant, which promotes a lower morbidity despite being much more contagious. In this perspective, we performed molecular dynamics (MD) simulations of the different spike RBD/hACE2 complexes corresponding to the WT, Delta and four Omicron variants. Carrying out a comprehensive analysis of residue interactions within and between the two partners allowed us to draw the profile of each variant by using complementary methods (PairInt, hydrophobic potential, contact PCA). PairInt calculations highlighted the residues most involved in electrostatic interactions, which make a strong contribution to the binding with highly stable interactions between spike RBD and hACE2. Apolar contacts made a substantial and complementary contribution in Omicron with the detection of two hydrophobic patches. Contact networks and cross-correlation matrices were able to detect subtle changes at point mutations as the S375F mutation occurring in all Omicron variants, which is likely to confer an advantage in binding stability. This study brings new highlights on the dynamic binding of spike RBD to hACE2, which may explain the final persistence of Omicron over Delta.
Collapse
Affiliation(s)
- Aria Gheeraert
- Laboratory of Mathematics (LAMA), CNRS, University of Savoie Mont Blanc, 73370 Le Bourget-du-Lac, France; (A.G.); (L.V.)
- Dipartimento di Chimica Industriale “Toso Montanari”, Università di Bologna, Viale del Risorgimento, 40129 Bologna, Italy;
| | - Vincent Leroux
- LORIA, CNRS, Inria, University of Lorraine, 54506 Vandoeuvre-lès-Nancy, France; (V.L.); (D.M.-L.); (Y.K.); (I.C.d.B.); (M.-D.D.)
| | - Dominique Mias-Lucquin
- LORIA, CNRS, Inria, University of Lorraine, 54506 Vandoeuvre-lès-Nancy, France; (V.L.); (D.M.-L.); (Y.K.); (I.C.d.B.); (M.-D.D.)
| | - Yasaman Karami
- LORIA, CNRS, Inria, University of Lorraine, 54506 Vandoeuvre-lès-Nancy, France; (V.L.); (D.M.-L.); (Y.K.); (I.C.d.B.); (M.-D.D.)
| | - Laurent Vuillon
- Laboratory of Mathematics (LAMA), CNRS, University of Savoie Mont Blanc, 73370 Le Bourget-du-Lac, France; (A.G.); (L.V.)
| | - Isaure Chauvot de Beauchêne
- LORIA, CNRS, Inria, University of Lorraine, 54506 Vandoeuvre-lès-Nancy, France; (V.L.); (D.M.-L.); (Y.K.); (I.C.d.B.); (M.-D.D.)
| | - Marie-Dominique Devignes
- LORIA, CNRS, Inria, University of Lorraine, 54506 Vandoeuvre-lès-Nancy, France; (V.L.); (D.M.-L.); (Y.K.); (I.C.d.B.); (M.-D.D.)
| | - Ivan Rivalta
- Dipartimento di Chimica Industriale “Toso Montanari”, Università di Bologna, Viale del Risorgimento, 40129 Bologna, Italy;
- ENS, CNRS, Laboratoire de Chimie UMR 5182, 69364 Lyon, France
| | - Bernard Maigret
- LORIA, CNRS, Inria, University of Lorraine, 54506 Vandoeuvre-lès-Nancy, France; (V.L.); (D.M.-L.); (Y.K.); (I.C.d.B.); (M.-D.D.)
| | - Laurent Chaloin
- Institut de Recherche en Infectiologie de Montpellier (IRIM), CNRS, University of Montpellier, 34293 Montpellier, France
| |
Collapse
|
8
|
Lotthammer JM, Holehouse AS. Disentangling Folding from Energetic Traps in Simulations of Disordered Proteins. J Chem Inf Model 2025; 65:2897-2910. [PMID: 40042172 DOI: 10.1021/acs.jcim.4c02005] [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: 03/25/2025]
Abstract
Protein conformational heterogeneity plays an essential role in a myriad of different biological processes. Extensive conformational heterogeneity is especially characteristic of intrinsically disordered proteins and protein regions (collectively IDRs), which lack a well-defined three-dimensional structure and instead rapidly exchange between a diverse ensemble of configurations. An emerging paradigm recognizes that the conformational biases encoded in IDR ensembles can play a central role in their biological function, necessitating understanding these sequence-ensemble relations. All-atom simulations have provided critical insight into our modern understanding of the solution behavior of IDRs. However, effectively exploring the accessible conformational space associated with large, heterogeneous ensembles is challenging. In particular, identifying poorly sampled or energetically trapped regions of disordered proteins in simulations often relies on qualitative assessment based on visual inspection of simulations and/or analysis data. These approaches, while convenient, run the risk of masking poorly sampled simulations. In this work, we present an algorithm for quantifying per-residue local conformational heterogeneity in protein simulations. Our work builds on prior work and compares the similarity between backbone dihedral angle distributions generated from molecular simulations in a limiting polymer model and across independent all-atom simulations. In this regime, the polymer model serves as a statistical reference model for extensive conformational heterogeneity in a real chain. Quantitative comparisons of probability vectors generated from these simulations reveal the extent of conformational sampling in a simulation, enabling us to distinguish between situations in which protein regions are well-sampled, poorly sampled, or folded. To demonstrate the effectiveness of this approach, we apply our algorithm to several toy, synthetic, and biological systems. Accurately assessing local conformational sampling in simulations of IDRs will help better quantify new enhanced sampling methods, ensure force field comparisons are equivalent, and provide confidence that conclusions drawn from simulations are robust.
Collapse
Affiliation(s)
- Jeffrey M Lotthammer
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri 63110, United States
- Center for Biomolecular Condensates (CBC), Washington University in St. Louis, St. Louis, Missouri 63110, United States
| | - Alex S Holehouse
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, Missouri 63110, United States
- Center for Biomolecular Condensates (CBC), Washington University in St. Louis, St. Louis, Missouri 63110, United States
| |
Collapse
|
9
|
Vaiani L, Uva AE, Boccaccio A. Lattice Models: Non-Conventional simulation methods for mechanobiology. J Biomech 2025; 181:112555. [PMID: 39892284 DOI: 10.1016/j.jbiomech.2025.112555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 12/30/2024] [Accepted: 01/23/2025] [Indexed: 02/03/2025]
Abstract
Computational methods represent a powerful tool to explore biophysical phenomena occurring at small scales and hence difficult to observe through experimental setups. In detail, they can provide a support to mechanobiology, with the aim of understanding the behavior of living cells interacting with the surrounding environment. To this end, lattice models can provide a simulation framework that is highly reliable and easy to implement, even for simulations involving large deformations and topological changes during time evolution. In this review article, elastic network models for studying biological molecules are described, several lattice spring models for investigating cell behaviors are discussed, and the adoption of lattice beam models for biomimetic structures design is presented. The lattice modelling approaches could be regarded as a valuable option to conduct in-silico experiments and consolidate the emergent mechanobiology research field.
Collapse
Affiliation(s)
- Lorenzo Vaiani
- Dipartimento di Meccanica, Matematica e Management, Politecnico di Bari, Via Orabona, 4, 70125 Bari, Italy.
| | - Antonio Emmanuele Uva
- Dipartimento di Meccanica, Matematica e Management, Politecnico di Bari, Via Orabona, 4, 70125 Bari, Italy
| | - Antonio Boccaccio
- Dipartimento di Meccanica, Matematica e Management, Politecnico di Bari, Via Orabona, 4, 70125 Bari, Italy.
| |
Collapse
|
10
|
Mittal J, Phan T, Mohanty P. Optimal scaling of protein-water interactions coupled with targeted torsional refinements yields balanced force fields suitable for simulations of single-chain folded proteins, disordered polypeptides, and protein-protein complexes. RESEARCH SQUARE 2025:rs.3.rs-5932820. [PMID: 40060049 PMCID: PMC11888540 DOI: 10.21203/rs.3.rs-5932820/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
All-atom molecular dynamics (MD) simulations based on physics-based force fields, serve as an essential complement to experiments for investigating protein structure, dynamics, and interactions. Despite significant advances in force field development, achieving a consistent balance of molecular interactions that stabilize folded proteins and protein-protein complexes while simultaneously capturing the conformational dynamics of intrinsically disordered polypeptides (IDPs), remains challenging. In this work, we systematically evaluated two current state-of-the-art force fields (i) AMBER ff03ws, and (ii) AMBER ff99SBws, by comprehensively assessing their performance on both folded domains and IDPs. By selectively scaling side chain-water interactions for uncharged residues, the refined AMBER ff03w-sc force field demonstrated improved conformational stability of folded proteins while maintaining accurate representations of IDPs. However, AMBER ff03w-sc failed to correct the discrepancies in NMR-derived ps-ns timescale backbone dynamics associated with flexible loops. Interestingly, AMBER ff99SBws retained its structural stability despite the application of upscaled interactions with water for both sidechain and backbone atoms and displayed robust agreement with NMR-derived backbone dynamics. Further, a targeted refinement of glutamine backbone torsion parameters, yielded AMBER ff99SBws-STQ', which effectively resolved discrepancies associated with glutamine α-helicity predictions. Extensive validation against small angle X-ray scattering (SAXS) and NMR chemical shifts, revealed that both refined force fields accurately reproduced chain dimensions and secondary structure propensities of disordered peptides and prion-like domains. Importantly, both force fields reliably maintained the stability of protein-protein complexes over microsecond timescales. Our systematic refinement strategies provide improved accuracy and transferability for simulating diverse protein systems, from folded domains to IDPs and protein complexes.
Collapse
|
11
|
Zhao F, Mao Y, Yang J, Yang S, Guan X, Wang Z, Huang T. Enhancing Bacillus thuringiensis Performance: Fertilizer-Driven Improvements in Biofilm Formation, UV Protection, and Pest Control Efficacy. Microorganisms 2025; 13:499. [PMID: 40142392 PMCID: PMC11945023 DOI: 10.3390/microorganisms13030499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
This study investigated the effects of fertilizers on the biofilm formation, ultraviolet (UV) resistance, and insecticidal activity of Bacillus thuringiensis (Bt). Bacillus thuringiensis, a widely used microbial pesticide, has a minimal environmental impact and is highly effective against specific pests but is susceptible to environmental factors in field applications. Bacterial biofilms provide protection for Bt, enhancing its survival and functionality in the environment. However, the mechanisms by which fertilizers regulate the characteristics of microbial pesticides and enhance biofilm formation are not well understood. This study evaluated the effects of six fertilizers on the bacterial biofilm formation, the UV resistance, and the insecticidal activities of Bt wettable powders. The results demonstrated that fertilizers significantly enhanced the performance of three Bt preparations (Lv'an, Kang'xin, and Lu'kang). A compound fertilizer with 8.346 g/L of KCl, 2.751 g/L of ZnSO4·7H2O, and 25.681 μL/mL of humic acid was identified by response surface optimization, achieving the maximum BBF formation with OD595 value of 2.738. Furthermore, KH2PO4, HA, and ZnSO4·7H2O notably improved the survivability of Bt preparations under prolonged UV exposure, with the compound fertilizer providing the greatest protection. What's more, fertilizers reduced the LC50 values of all Bt preparations, with the compound fertilizer decreasing the LC50 of the Lv'an Bt wettable powder to 0.139 g/L, a 3.12-fold increase in efficacy. This study demonstrated that fertilizers significantly enhance the UV resistance and insecticidal activity of Bt wettable powders by promoting bacterial biofilm formation. Herein, this study provides new strategies and theoretical support for Bt applications in modern sustainable agriculture.
Collapse
Affiliation(s)
| | | | | | | | | | - Zixuan Wang
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Key Laboratory of Biopesticide and Chemical Biology of Ministry of Education, Biopesticide Research Center, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (F.Z.); (Y.M.); (J.Y.); (S.Y.); (X.G.)
| | - Tianpei Huang
- State Key Laboratory of Ecological Pest Control for Fujian and Taiwan Crops, Key Laboratory of Biopesticide and Chemical Biology of Ministry of Education, Biopesticide Research Center, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (F.Z.); (Y.M.); (J.Y.); (S.Y.); (X.G.)
| |
Collapse
|
12
|
Atanasova M. Small-Molecule Inhibitors of Amyloid Beta: Insights from Molecular Dynamics-Part A: Endogenous Compounds and Repurposed Drugs. Pharmaceuticals (Basel) 2025; 18:306. [PMID: 40143085 PMCID: PMC11944459 DOI: 10.3390/ph18030306] [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: 02/10/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
The amyloid hypothesis is the predominant model of Alzheimer's disease (AD) pathogenesis, suggesting that amyloid beta (Aβ) peptide is the primary driver of neurotoxicity and a cascade of pathological events in the central nervous system. Aβ aggregation into oligomers and deposits triggers various processes, such as vascular damage, inflammation-induced astrocyte and microglia activation, disrupted neuronal ionic homeostasis, oxidative stress, abnormal kinase and phosphatase activity, tau phosphorylation, neurofibrillary tangle formation, cognitive dysfunction, synaptic loss, cell death, and, ultimately, dementia. Molecular dynamics (MD) is a powerful structure-based drug design (SBDD) approach that aids in understanding the properties, functions, and mechanisms of action or inhibition of biomolecules. As the only method capable of simulating atomic-level internal motions, MD provides unique insights that cannot be obtained through other techniques. Integrating experimental data with MD simulations allows for a more comprehensive understanding of biological processes and molecular interactions. This review summarizes and evaluates MD studies from the past decade on small molecules, including endogenous compounds and repurposed drugs, that inhibit amyloid beta. Furthermore, it outlines key considerations for future MD simulations of amyloid inhibitors, offering a potential framework for studies aimed at elucidating the mechanisms of amyloid beta inhibition by small molecules.
Collapse
|
13
|
Das IJ, Bhatta K, Sarangi I, Samal HB. Innovative computational approaches in drug discovery and design. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2025; 103:1-22. [PMID: 40175036 DOI: 10.1016/bs.apha.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
In the current scenario of pandemics, drug discovery and design have undergone a significant transformation due to the integration of advanced computational methodologies. These methodologies utilize sophisticated algorithms, machine learning, artificial intelligence, and high-performance computing to expedite the drug development process, enhances accuracy, and reduces costs. Machine learning and AI have revolutionized predictive modeling, virtual screening, and de novo drug design, allowing for the identification and optimization of novel compounds with desirable properties. Molecular dynamics simulations provide a detailed insight into protein-ligand interactions and conformational changes, facilitating an understanding of drug efficacy at the atomic level. Quantum mechanics/molecular mechanics methods offer precise predictions of binding energies and reaction mechanisms, while structure-based drug design employs docking studies and fragment-based design to improve drug-receptor binding affinities. Network pharmacology and systems biology approaches analyze polypharmacology and biological networks to identify novel drug targets and understand complex interactions. Cheminformatics explores vast chemical spaces and employs data mining to find patterns in large datasets. Computational toxicology predicts adverse effects early in development, reducing reliance on animal testing. Bioinformatics integrates genomic, proteomic, and metabolomics data to discover biomarkers and understand genetic variations affecting drug response. Lastly, cloud computing and big data technologies facilitate high-throughput screening and comprehensive data analysis. Collectively, these computational innovations are driving a paradigm shift in drug discovery and design, making it more efficient, accurate, and cost-effective.
Collapse
Affiliation(s)
- Itishree Jogamaya Das
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Kalpita Bhatta
- Department of Botany, School of Applied Sciences, Centurion University of Technology and Management, Bhubaneswar, Odisha, India
| | - Itisam Sarangi
- Biomedical Engineering Department, University of Michigan, Ann Arbor, MI, United States
| | - Himansu Bhusan Samal
- School of Pharmacy and Life Sciences, Centurion University of Technology Management, Bhubaneswar, Odisha, India.
| |
Collapse
|
14
|
Ilter M, Escorcia AM, Schulze-Niemand E, Naumann M, Stein M. Activation and Reactivity of the Deubiquitinylase OTU Cezanne-2 from MD Simulations and QM/MM Calculations. J Chem Inf Model 2025; 65:921-936. [PMID: 39782030 PMCID: PMC11776055 DOI: 10.1021/acs.jcim.4c01964] [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: 10/24/2024] [Revised: 12/12/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025]
Abstract
Cezanne-2 (Cez2) is a deubiquitinylating (DUB) enzyme involved in the regulation of ubiquitin-driven cellular signaling and selectively targets Lys11-linked polyubiquitin chains. As a representative member of the ovarian tumor (OTU) subfamily DUBs, it performs cysteine proteolytic isopeptide bond cleavage; however, its exact catalytic mechanism is not yet resolved. In this work, we used different computational approaches to get molecular insights into the Cezanne-2 catalytic mechanism. Extensive molecular dynamics (MD) simulations were performed for 12 μs to model free Cez2 and the diubiquitin (diUb) substrate-bound protein-protein complex in two different charge states of Cez2, each corresponding to a distinct reactive state in its catalytic cycle. The simulations were analyzed in terms of the relevant structural parameters for productive enzymatic catalysis. Reactive diUb-Cez2 complex configurations were identified, which lead to isopeptide bond cleavage and stabilization of the tetrahedral oxyanion intermediate. The reliability of these complexes was further assessed by quantum mechanics/molecular mechanics (QM/MM) optimizations. The results show that Cez2 follows a modified cysteine protease mechanism involving a catalytic Cys210/His367 dyad, with the oxyanion hole to be a part of the "C-loop," and polarization of His367 by the formation of a strictly conserved water bridge with Glu173. The third residue has a dual role in catalysis as it mediates substrate binding and polarization of the catalytic dyad. A similar mechanism was identified for Cezanne-1, the paralogue of Cez2. In general, our simulations provide valuable molecular information that may help in the rational design of selective inhibitors of Cez2 and closely related enzymes.
Collapse
Affiliation(s)
- Metehan Ilter
- Molecular
Simulations and Design Group, Max Planck
Institute for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106 Magdeburg, Germany
| | - Andrés M. Escorcia
- Molecular
Simulations and Design Group, Max Planck
Institute for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106 Magdeburg, Germany
| | - Eric Schulze-Niemand
- Molecular
Simulations and Design Group, Max Planck
Institute for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106 Magdeburg, Germany
- Institute
for Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany
| | - Michael Naumann
- Institute
for Experimental Internal Medicine, Medical Faculty, Otto von Guericke University, Leipziger Straße 44, 39120 Magdeburg, Germany
| | - Matthias Stein
- Molecular
Simulations and Design Group, Max Planck
Institute for Dynamics of Complex Technical Systems, Sandtorstrasse 1, 39106 Magdeburg, Germany
| |
Collapse
|
15
|
Chan MC, Alfawaz Y, Paul A, Shukla D. Molecular insights into the elevator-type mechanism of the cyanobacterial bicarbonate transporter BicA. Biophys J 2025; 124:379-392. [PMID: 39674889 PMCID: PMC11788499 DOI: 10.1016/j.bpj.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 03/17/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024] Open
Abstract
Cyanobacteria are responsible for up to 80% of aquatic carbon dioxide fixation and have evolved a specialized carbon concentrating mechanism to increase photosynthetic yield. As such, cyanobacteria are attractive targets for synthetic biology and engineering approaches to address the demands of global energy security, food production, and climate change for an increasing world's population. The bicarbonate transporter BicA is a sodium-dependent, low-affinity, high-flux bicarbonate symporter expressed in the plasma membrane of cyanobacteria. Despite extensive biochemical characterization of BicA, including the resolution of the BicA crystal structure, the dynamic understanding of the bicarbonate transport mechanism remains elusive. To this end, we have collected over 1 ms of all-atom molecular dynamics simulation data of the BicA dimer to elucidate the structural rearrangements involved in the substrate transport process. We further characterized the energetics of the transition of BicA protomers and investigated potential mutations that are shown to decrease the free energy barrier of conformational transitions. In all, our study illuminates a detailed mechanistic understanding of the conformational dynamics of bicarbonate transporters and provides atomistic insights to engineering these transporters for enhanced photosynthetic production.
Collapse
Affiliation(s)
- Matthew C Chan
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Yazeed Alfawaz
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Arnav Paul
- Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Diwakar Shukla
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois; Department of Chemistry, University of Illinois Urbana-Champaign, Urbana, Illinois; Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, Illinois; Department of Plant Biology, University of Illinois Urbana-Champaign, Urbana, Illinois; Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois.
| |
Collapse
|
16
|
Wang X, Xiong D, Zhang Y, Zhai J, Gu YC, He X. The evolution of the Amber additive protein force field: History, current status, and future. J Chem Phys 2025; 162:030901. [PMID: 39817575 DOI: 10.1063/5.0227517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/30/2024] [Indexed: 01/18/2025] Open
Abstract
Molecular dynamics simulations are pivotal in elucidating the intricate properties of biological molecules. Nonetheless, the reliability of their outcomes hinges on the precision of the molecular force field utilized. In this perspective, we present a comprehensive review of the developmental trajectory of the Amber additive protein force field, delving into researchers' persistent quest for higher precision force fields and the prevailing challenges. We detail the parameterization process of the Amber protein force fields, emphasizing the specific improvements and retained features in each version compared to their predecessors. Furthermore, we discuss the challenges that current force fields encounter in balancing the interactions of protein-protein, protein-water, and water-water in molecular dynamics simulations, as well as potential solutions to overcome these issues.
Collapse
Affiliation(s)
- Xianwei Wang
- School of Physics, Zhejiang University of Technology, Hangzhou, Zhejiang 310023, China
| | - Danyang Xiong
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Shanghai Frontiers Science Center of Molecule Intelligent Syntheses, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Yueqing Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Shanghai Frontiers Science Center of Molecule Intelligent Syntheses, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Jihang Zhai
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Shanghai Frontiers Science Center of Molecule Intelligent Syntheses, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Yu-Cheng Gu
- Syngenta Jealott's Hill International Research Centre Bracknell, Berkshire RG42 6EY, United Kingdom
| | - Xiao He
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Shanghai Frontiers Science Center of Molecule Intelligent Syntheses, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
- Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing 401120, China
- New York University-East China Normal University Center for Computational Chemistry, New York University Shanghai, Shanghai 200062, China
| |
Collapse
|
17
|
Stegani B, Scalone E, Toplek FB, Löhr T, Gianni S, Vendruscolo M, Capelli R, Camilloni C. Estimation of Ligand Binding Free Energy Using Multi-eGO. J Chem Inf Model 2025; 65:351-362. [PMID: 39737687 DOI: 10.1021/acs.jcim.4c01545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2025]
Abstract
The computational study of ligand binding to a target protein provides mechanistic insight into the molecular determinants of this process and can improve the success rate of in silico drug design. All-atom molecular dynamics (MD) simulations can be used to evaluate the binding free energy, typically by thermodynamic integration, and to probe binding mechanisms, including the description of protein conformational dynamics. The advantages of MD come at a high computational cost, which limits its use. Such cost could be reduced by using coarse-grained models, but their use is generally associated with an undesirable loss of resolution and accuracy. To address the trade-off between speed and accuracy of MD simulations, we describe the use of the recently introduced multi-eGO atomic model for the estimation of binding free energies. We illustrate this approach in the case of the binding of benzene to lysozyme by both thermodynamic integration and metadynamics, showing multiple binding/unbinding pathways of benzene. We then provide equally accurate results for the binding free energy of dasatinib and PP1 to Src kinase by thermodynamic integration. Finally, we show how we can describe the binding of the small molecule 10074-G5 to Aβ42 by single molecule simulations and by explicit titration of the ligand as a function of concentration. These results demonstrate that multi-eGO has the potential to significantly reduce the cost of accurate binding free energy calculations and can be used to develop and benchmark in silico ligand binding techniques.
Collapse
Affiliation(s)
- Bruno Stegani
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan 20133, Italy
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Istituto Pasteur-Fondazione Cenci Bolognetti and Istituto di Biologia e Patologia Molecolari del CNR, Sapienza Università di Roma, Rome 00185, Italy
| | - Emanuele Scalone
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan 20133, Italy
- Department of Chemistry, Dartmouth College, Hanover, New Hampshire 03755, United States
| | - Fran Bačić Toplek
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan 20133, Italy
| | - Thomas Löhr
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| | - Stefano Gianni
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Istituto Pasteur-Fondazione Cenci Bolognetti and Istituto di Biologia e Patologia Molecolari del CNR, Sapienza Università di Roma, Rome 00185, Italy
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, U.K
| | - Riccardo Capelli
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan 20133, Italy
| | - Carlo Camilloni
- Dipartimento di Bioscienze, Università degli Studi di Milano, Milan 20133, Italy
| |
Collapse
|
18
|
Roy A, Ward E, Choi I, Cosi M, Edgin T, Hughes TS, Islam MS, Khan AM, Kolekar A, Rayl M, Robinson I, Sarando P, Skidmore E, Swetnam TL, Wall M, Xu Z, Yung M, Merchant N, Wheeler TJ. MDRepo-an open data warehouse for community-contributed molecular dynamics simulations of proteins. Nucleic Acids Res 2025; 53:D477-D486. [PMID: 39535038 PMCID: PMC11701643 DOI: 10.1093/nar/gkae1109] [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: 09/13/2024] [Revised: 10/15/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Molecular Dynamics (MD) simulation of biomolecules provides important insights into conformational changes and dynamic behavior, revealing critical information about folding and interactions with other molecules. The collection of simulations stored in computers across the world holds immense potential to serve as training data for future Machine Learning models that will transform the prediction of structure, dynamics, drug interactions, and more. Ideally, there should exist an open access repository that enables scientists to submit and store their MD simulations of proteins and protein-drug interactions, and to find, retrieve, analyze, and visualize simulations produced by others. However, despite the ubiquity of MD simulation in structural biology, no such repository exists; as a result, simulations are instead stored in scattered locations without uniform metadata or access protocols. Here, we introduce MDRepo, a robust infrastructure that provides a relatively simple process for standardized community contribution of simulations, activates common downstream analyses on stored data, and enables search, retrieval, and visualization of contributed data. MDRepo is built on top of the open-source CyVerse research cyber-infrastructure, and is capable of storing petabytes of simulations, while providing high bandwidth upload and download capabilities and laying a foundation for cloud-based access to its stored data.
Collapse
Affiliation(s)
- Amitava Roy
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA
| | - Ethan Ward
- R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | | | - Michele Cosi
- Data Science Institute, University of Arizona, Tucson, AZ, USA
| | - Tony Edgin
- CyVerse, University of Arizona, Tucson, AZ, USA
| | - Travis S Hughes
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA
- Biochemistry and Biophysics, University of Montana, Missoula, MT, USA
| | - Md Shafayet Islam
- Department of Physics, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Asif M Khan
- University of Doha for Science and Technology, Doha, Qatar
| | - Aakash Kolekar
- R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Mariah Rayl
- Biochemistry and Biophysics, University of Montana, Missoula, MT, USA
| | - Isaac Robinson
- R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | | | | | | | - Mariah Wall
- CyVerse, University of Arizona, Tucson, AZ, USA
| | - Zhuoyun Xu
- CyVerse, University of Arizona, Tucson, AZ, USA
| | | | | | - Travis J Wheeler
- R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
19
|
Rassier DE, Månsson A. Mechanisms of myosin II force generation: insights from novel experimental techniques and approaches. Physiol Rev 2025; 105:1-93. [PMID: 38451233 DOI: 10.1152/physrev.00014.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024] Open
Abstract
Myosin II is a molecular motor that converts chemical energy derived from ATP hydrolysis into mechanical work. Myosin II isoforms are responsible for muscle contraction and a range of cell functions relying on the development of force and motion. When the motor attaches to actin, ATP is hydrolyzed and inorganic phosphate (Pi) and ADP are released from its active site. These reactions are coordinated with changes in the structure of myosin, promoting the so-called "power stroke" that causes the sliding of actin filaments. The general features of the myosin-actin interactions are well accepted, but there are critical issues that remain poorly understood, mostly due to technological limitations. In recent years, there has been a significant advance in structural, biochemical, and mechanical methods that have advanced the field considerably. New modeling approaches have also allowed researchers to understand actomyosin interactions at different levels of analysis. This paper reviews recent studies looking into the interaction between myosin II and actin filaments, which leads to power stroke and force generation. It reviews studies conducted with single myosin molecules, myosins working in filaments, muscle sarcomeres, myofibrils, and fibers. It also reviews the mathematical models that have been used to understand the mechanics of myosin II in approaches focusing on single molecules to ensembles. Finally, it includes brief sections on translational aspects, how changes in the myosin motor by mutations and/or posttranslational modifications may cause detrimental effects in diseases and aging, among other conditions, and how myosin II has become an emerging drug target.
Collapse
Affiliation(s)
- Dilson E Rassier
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, Canada
| | - Alf Månsson
- Physiology, Linnaeus University, Kalmar, Sweden
| |
Collapse
|
20
|
Yu Y, Weng J, Wang W. Use of Steered Molecular Dynamics to Explore the Conformational Changes of SNARE Proteins. Methods Mol Biol 2025; 2887:69-77. [PMID: 39806146 DOI: 10.1007/978-1-0716-4314-3_4] [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: 01/16/2025]
Abstract
Steered Molecular Dynamics (SMD) simulation is a powerful computational simulation technique that enables the controlled manipulation of molecular systems by applying external forces. This method is frequently utilized to investigate the slow processes of biomolecular systems that occur within sub-second to second time scales, achieved through SMD simulations that only span nanoseconds. SMD simulation can be utilized to study the detailed mechanism of protein conformational changes, protein unfolding, and ligand dissociation, etc. Here, we introduce the application of SMD in the study of intra-molecular interactions and regulatory mechanisms in SNAREs by analyzing their conformational change pathways.
Collapse
Affiliation(s)
- Yiping Yu
- Department of Chemistry, Institute of Biomedical Sciences and Multiscale Research Institute of Complex Systems, Fudan University, Shanghai, China
| | - Jingwei Weng
- Department of Chemistry, Institute of Biomedical Sciences and Multiscale Research Institute of Complex Systems, Fudan University, Shanghai, China
| | - Wenning Wang
- Department of Chemistry, Institute of Biomedical Sciences and Multiscale Research Institute of Complex Systems, Fudan University, Shanghai, China.
| |
Collapse
|
21
|
Wang D, Tiwary P. Augmenting Human Expertise in Weighted Ensemble Simulations through Deep Learning-Based Information Bottleneck. J Chem Theory Comput 2024; 20:10371-10383. [PMID: 39589127 DOI: 10.1021/acs.jctc.4c00919] [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: 11/27/2024]
Abstract
The weighted ensemble (WE) method stands out as a widely used segment-based sampling technique renowned for its rigorous treatment of kinetics. The WE framework typically involves initially mapping the configuration space onto a low-dimensional collective variable (CV) space and then partitioning it into bins. The efficacy of WE simulations heavily depends on the selection of CVs and binning schemes. The recently proposed state predictive information bottleneck (SPIB) method has emerged as a promising tool for automatically constructing CVs from data and guiding enhanced sampling through an iterative manner. In this work, we advance this data-driven pipeline by incorporating prior expert knowledge. Our hybrid approach combines SPIB-learned CVs to enhance sampling in explored regions with expert-based CVs to guide exploration in regions of interest, synergizing the strengths of both methods. Through benchmarking on alanine dipeptide and chignolin systems, we demonstrate that our hybrid approach effectively guides WE simulations to sample states of interest and reduces run-to-run variances. Moreover, our integration of the SPIB model also enhances the analysis and interpretation of WE simulation data by effectively identifying metastable states and pathways and offering direct visualization of dynamics.
Collapse
Affiliation(s)
- Dedi Wang
- Biophysics Program and Institute for Physical Science and Technology, University of Maryland, College Park, Maryland 20742, United States
| | - Pratyush Tiwary
- Department of Chemistry and Biochemistry and Institute for Physical Science and Technology, University of Maryland, College Park, Maryland 20742, United States
- University of Maryland Institute for Health Computing, Bethesda, Maryland 20852, United States
| |
Collapse
|
22
|
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] [Download PDF] [Figures] [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
|
23
|
Abdalla M, Abdelkhalig SM, Edet UO, Zothantluanga JH, Umoh EA, Moglad E, Nkang NA, Hader MM, Alanazi TMR, AlShouli S, Al-Shouli S. Molecular dynamics-based computational investigations on the influence of tumor suppressor p53 binding protein against other proteins/peptides. Sci Rep 2024; 14:29871. [PMID: 39622863 PMCID: PMC11612205 DOI: 10.1038/s41598-024-81499-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024] Open
Abstract
The tumor-suppressing p-53 binding protein is a crucial protein that is involved in the prevention of cancer via its regulatory effect on a number of cellular processes. Recent evidence indicates that it interacts with a number of other proteins involved in cancer in ways that are not fully understood. An understanding of such interactions could provide insights into novel ways p53 further exerts its tumour prevention role via its interactions with diverse proteins. Thus, this study aimed to examine the interactions of the p53 protein with other proteins (peptides and histones) using molecular simulation dynamics. We opted for a total of seven proteins, namely 2LVM, 2MWO, 2MWP, 4CRI, 4 × 34, 5Z78, and 6MYO (control), and had their PBD files retrieved from the protein database. These proteins were then docked against the p-53 protein and the resulting interactions were examined using molecular docking simulations run at 500 ns. The result of the interactions revealed the utilisation of various amino acids in the process. The peptide that interacted with the highest number of amino acids was 5Z78 and these were Lys10, Gly21, Trp24, Pro105, His106, and Arg107, indicating a stronger interaction. The RMSD and RMSF values indicate that the complexes formed were stable, with 4CRI, 6MYO, and 2G3R giving the most stable values (less than 2.5 Å). Other parameters, including the SASA, Rg, and number of hydrogen bonds, all indicated the formation of fairly stable complexes. Our study indicates that overall, the interactions of 53BP1 with p53K370me2, p53K382me2, methylated K810 Rb, p53K381acK382me2, and tudor-interacting repair regulator protein indicated interactions that were not as strong as those with the histone protein. Thus, it could be that P53 may mediate its tumour suppressing effect via interactions with amino acids and histone.
Collapse
Affiliation(s)
- Mohnad Abdalla
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University, Jinan, China.
| | - Sozan M Abdelkhalig
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh, 11597, Saudi Arabia
| | - Uwem O Edet
- Department of Biological (Microbiology), Faculty of Natural and Applied Sciences, Arthur Jarvis University, Akpabuyo, Cross River State, Nigeria.
| | - James H Zothantluanga
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Ekementeabasi Aniebo Umoh
- Department of Human Physiology, Faculty of Basic Medical Sciences, Arthur Jarvis University, Akpabuyo, Cross River State, Nigeria
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, P.O. Box 173, Alkharj, 11942, Saudi Arabia
| | - Nkoyo Ani Nkang
- Science Laboratory Department, Faculty of Biological Sciences, University of Calabar, Calabar, Cross River State, Nigeria
| | - Meshari M Hader
- Dietary Department, Dr. Soliman Fakeeh Hospital, Jeddah, Saudi Arabia
| | | | - Sawsan AlShouli
- Pharmacy Department, Security Forces Hospital, Riyadh, 11481, Saudi Arabia
| | - Samia Al-Shouli
- Immunology Unit, Department of Pathology, College of Medicine, King Saud University, Riyadh, 11461, Saudi Arabia
| |
Collapse
|
24
|
Sahrmann P, Voth GA. Enhancing the Assembly Properties of Bottom-Up Coarse-Grained Phospholipids. J Chem Theory Comput 2024; 20:10235-10246. [PMID: 39535391 PMCID: PMC11604101 DOI: 10.1021/acs.jctc.4c00905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/06/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
A plethora of key biological events occur at the cellular membrane where the large spatiotemporal scales necessitate dimensionality reduction or coarse-graining approaches over conventional all-atom molecular dynamics simulation. Constructing coarse-grained descriptions of membranes systematically from statistical mechanical principles has largely remained challenging due to the necessity of capturing amphipathic self-assembling behavior in coarse-grained models. We show that bottom-up coarse-grained lipid models can possess metastable morphological behavior and that this potential metastability has ramifications for accurate development and training. We in turn develop a training algorithm which evades metastability issues by linking model training to self-assembling behavior, and demonstrate its robustness via construction of solvent-free coarse-grained models of various phospholipid membranes, including lipid species such as phosphatidylcholines, phosphatidylserines, sphingolipids, and cholesterol. The resulting coarse-grained lipid models are orders of magnitude faster than their atomistic counterparts while retaining structural fidelity and constitute a promising direction for the development of coarse-grained models of realistic cell membranes.
Collapse
Affiliation(s)
- Patrick
G. Sahrmann
- Department of Chemistry,
Chicago Center for Theoretical Chemistry, James Franck Institute,
and Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois 60637, United States
| | - Gregory A. Voth
- Department of Chemistry,
Chicago Center for Theoretical Chemistry, James Franck Institute,
and Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois 60637, United States
| |
Collapse
|
25
|
Chauhan K, Vijayan V, Pant P, Sharma S, Hassan MI, Ganguly NK, Sharma P, Rana R. Identifying potential inhibitors against galectin-3-binding protein (LGALS3BP) for therapeutic targeting of glioma. J Biomol Struct Dyn 2024:1-13. [PMID: 39589107 DOI: 10.1080/07391102.2024.2431185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 04/18/2024] [Indexed: 11/27/2024]
Abstract
Glioblastoma is one of the most lethal types of Gliomas, and its treatment greatly depends on the stage of its detection. Galactin-3-binding protein (LGALS3BP) serves as a novel circulatory biomarker for detecting glioma at an early stage. This protein is also responsible for metastasis, proliferative signaling, angiogenesis, and immune system evasion in the case of brain tumors. Inhibition of LGALS3BP can help in the reduction of metastasis and progression of the disease. Currently, no effective drug is available that can completely treat glioma. In this study, we have virtually screened the National Cancer Institute (NCI) drug databank to discover potential inhibitors of LGALS3BP. Based on the binding free energy calculations using MMPBSA, three compounds, 627861 (-16.69 kcal/mol), 329090 (-13.66 kcal/mol), and 627855 (-10.01 kcal/mol), were selected as potent inhibitors. 200 ns MD simulation studies further complemented this study. Finally, we recommend three molecules, 627861, 329090, and 627855, can be potential inhibitors of LGAL3SBP. The structural scaffolds of these molecules can also lead to the optimization of better inhibitors of LGALS3BP and be implicated in the therapeutic management of glioma after desired experimental validations.
Collapse
Affiliation(s)
- Kirti Chauhan
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi, India
| | - Viswanathan Vijayan
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Pradeep Pant
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, India
| | - Sujata Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Nirmal Kumar Ganguly
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi, India
| | - Pradeep Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Rashmi Rana
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi, India
| |
Collapse
|
26
|
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] [Grants] [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
|
27
|
Alghamdi A, A Awadh Ali N, Alafnan A, Zainal Abidin SA, Alamri A, Hussein W, Khalifa NE, Awaji AYM, Al Rashah K, Ahmed Younes KM, Anwar S. Deciphering the toxicological and computational assessment of Verbascum yemenese.: Integrating phytochemistry and bioinformatics tools. Food Chem Toxicol 2024; 193:115028. [PMID: 39368542 DOI: 10.1016/j.fct.2024.115028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 09/17/2024] [Accepted: 09/29/2024] [Indexed: 10/07/2024]
Abstract
This study explores the phytochemical composition and biological activities of Verbascum yemenense, a plant known for its medicinal properties. The plant extract revealed a rich presence of bioactive compounds that exhibited significant antioxidant properties against free radicals. The enzyme inhibition potential was particularly notable against cholinesterases (AChE: 2.56 mg GALAE/g; BChE: 1.98 mg GALAE/g), and tyrosinase (87.94 mg KAE/g), α-glucosidase suggesting potential therapeutic applications in neurodegenerative diseases, skin disorders and diabetes. Molecular docking studies and Molecular Dynamics simulations, providing insights into the interaction mechanisms of the identified compounds with the target proteins. Molecular docking studies revealed high binding affinities of the phytoconstituents, with compounds like VY4 and phyllanthusol-A (VY15) showing substantial docking scores against AChE (-9.840 kcal/mol) and BChE (-9.853 kcal/mol), respectively. For instance, the RMSD values during the MD simulations for compound VY17 in the AML complex showed a stable conformation, fluctuating within a range of 0.75 Å to 1.75 Å, indicating a strong and consistent interaction with the enzyme. MESP studies highlighted VY17's distinctive electrostatic features, notably a pronounced electronegative region, which might contribute to its binding efficiency. These findings suggest that V. yemenense is a promising candidate for developing novel therapeutic agents.
Collapse
Affiliation(s)
- Adel Alghamdi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Baha University, P.O. Box 1988, Al-Baha, Saudi Arabia
| | - Nasser A Awadh Ali
- Department of Pharmacognosy and Medicinal Herbs, Faculty of Pharmacy, Al-Baha University, P.O. Box 1988, Al-Baha, Saudi Arabia
| | - Ahmed Alafnan
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Ha'il, Ha'il, 55476, Saudi Arabia
| | - Syafiq Asnawi Zainal Abidin
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Malaysia
| | - Abdulwahab Alamri
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Ha'il, Ha'il, 55476, Saudi Arabia
| | - Weiam Hussein
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Ha'il, Ha'il, 55476, Saudi Arabia
| | - Nasrin E Khalifa
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, Ha'il, 55476, Saudi Arabia
| | | | - Khaled Al Rashah
- Medical Services, Ministry of Interior, Comprehensive Specialized Clinic for Security Forces, Najran, Saudi Arabia
| | - Kareem Mahmoud Ahmed Younes
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Ha'il, Ha'il, 55476, Saudi Arabia; Department of Analytical Chemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Sirajudheen Anwar
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Ha'il, Ha'il, 55476, Saudi Arabia.
| |
Collapse
|
28
|
Gupta A, Ma H, Ramanathan A, Zerze GH. A Deep Learning-Driven Sampling Technique to Explore the Phase Space of an RNA Stem-Loop. J Chem Theory Comput 2024; 20:9178-9189. [PMID: 39374435 DOI: 10.1021/acs.jctc.4c00669] [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: 10/09/2024]
Abstract
The folding and unfolding of RNA stem-loops are critical biological processes; however, their computational studies are often hampered by the ruggedness of their folding landscape, necessitating long simulation times at the atomistic scale. Here, we adapted DeepDriveMD (DDMD), an advanced deep learning-driven sampling technique originally developed for protein folding, to address the challenges of RNA stem-loop folding. Although tempering- and order parameter-based techniques are commonly used for similar rare-event problems, the computational costs or the need for a priori knowledge about the system often present a challenge in their effective use. DDMD overcomes these challenges by adaptively learning from an ensemble of running MD simulations using generic contact maps as the raw input. DeepDriveMD enables on-the-fly learning of a low-dimensional latent representation and guides the simulation toward the undersampled regions while optimizing the resources to explore the relevant parts of the phase space. We showed that DDMD estimates the free energy landscape of the RNA stem-loop reasonably well at room temperature. Our simulation framework runs at a constant temperature without external biasing potential, hence preserving the information on transition rates, with a computational cost much lower than that of the simulations performed with external biasing potentials. We also introduced a reweighting strategy for obtaining unbiased free energy surfaces and presented a qualitative analysis of the latent space. This analysis showed that the latent space captures the relevant slow degrees of freedom for the RNA folding problem of interest. Finally, throughout the manuscript, we outlined how different parameters are selected and optimized to adapt DDMD for this system. We believe this compendium of decision-making processes will help new users adapt this technique for the rare-event sampling problems of their interest.
Collapse
Affiliation(s)
- Ayush Gupta
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas 77204, United States
| | - Heng Ma
- Data Science and Learning Division, Argonne National Laboratory, Lemont, Illinois 60439, United States
| | - Arvind Ramanathan
- Data Science and Learning Division, Argonne National Laboratory, Lemont, Illinois 60439, United States
| | - Gül H Zerze
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas 77204, United States
| |
Collapse
|
29
|
Xu Z, Su P, Zhou X, Zheng Z, Zhu Y, Wang Q. Exploring the mechanism of action of Modified Simiao Powder in the treatment of osteoarthritis: an in-silico study. Front Med (Lausanne) 2024; 11:1422306. [PMID: 39493720 PMCID: PMC11527633 DOI: 10.3389/fmed.2024.1422306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024] Open
Abstract
Introduction Osteoarthritis (OA) is the most common form of arthritis and the leading musculoskeletal disorders in adults. Modified Simiao Powder (MSMP) has been widely used in the treatment of OA with remarkable clinical ecaciousness. Objective This study aimed to elucidate underlying mechanisms of MSMP in OA by employing network pharmacology, molecular docking, and molecular dynamics simulations, due to the unclear mode of action. Methods Bioinformatic analysis was used to evaluate the major chemical constituents of MSMP, determine prospective target genes, and screen genes associated with OA. Network pharmacology methods were then applied to identify the crucial target genes of MSMP in OA treatment. Further analyses included gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment. These key targets within the pertinent pathways was further confirmed by molecular docking, binding energy evaluation, and molecular dynamics simulations. Results Network pharmacology analysis identified an MSMP component-target-pathway network comprising 11 central active compounds, 25 gene targets, and 12 biological pathways. Discussion These findings imply that the therapeutic effects of MSMP was potentially mediated by targeting several pivotal genes, such as androgen receptor (AR), NFKB1, AKT1, MAPK1, and CASP3, and regulating some pathways, including lipid metabolism and atherosclerosis, the AGE-RAGE signaling pathway in diabetic complications, the PI3K-Akt signaling pathway, fluid shear stress, atherosclerosis, and Kaposi's sarcoma-associated herpesvirus infection. Molecular docking assessments demonstrated that these compounds of MSMP, such as berberine, kaempferol, quercetin, and luteolin, exhibit high binding anities to AR and AKT1. Molecular dynamics simulations validated the interactions between these compounds and targets. Conclusion The therapeutic effect of MSMP likely attributed to the modulation of multiple pathways, including lipid metabolism, atherosclerosis, the AGE-RAGE signaling pathway, and the PI3K-Akt signaling pathway, by the active components such as berberine, kaempferol, luteolin, and quercetin. Especially, their actions on target genes like AR and AKT1 contribute to the therapeutic benefits of MSMP observed in the treatment of OA.
Collapse
Affiliation(s)
- Zhouhengte Xu
- Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou, China
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Pingping Su
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiahui Zhou
- Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou, China
| | - Zhihui Zheng
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yibo Zhu
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qinglai Wang
- Wenzhou TCM Hospital of Zhejiang Chinese Medical University, Wenzhou, China
| |
Collapse
|
30
|
Yasuda T, Morita R, Shigeta Y, Harada R. BEMM-GEN: A Toolkit for Generating a Biomolecular Environment-Mimicking Model for Molecular Dynamics Simulation. J Chem Inf Model 2024; 64:7184-7188. [PMID: 39361452 PMCID: PMC11481083 DOI: 10.1021/acs.jcim.4c01467] [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: 08/16/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024]
Abstract
Understanding the influence of the cellular environment on protein conformations is crucial for elucidating protein functions within living cells. In studies using molecular dynamics (MD) simulation, carbon nanotubes and hydrophobic cages have been widely used to emulate the cellular environment inside specific large biomolecules such as ribosome tunnels and chaperones. However, recent studies suggest that these uniform hydrophobic models may not adequately capture the environmental effects inside each biomolecule. Based on these facts, it is necessary to generate spherical and cylindrical models with varied chemical properties corresponding to the components within target biomolecules. We developed a biomolecular environment-mimicking model generator (BEMM-GEN) that generates spherical and cylindrical models with user-specified chemical properties and allows the integration of arbitrary protein conformations into the generated models. BEMM-GEN provides model and protein complex structures, along with the corresponding parameter files for MD simulation (AMBER and GROMACS), and users immediately run their MD simulation based on the generated input files. BEMM-GEN can be freely downloaded and installed via a Python package manager (pip install BEMM-gen). The source code files and a user manual for operation are provided on GitHub (https://github.com/y4suda/BEMM-GEN).
Collapse
Affiliation(s)
- Takunori Yasuda
- Doctoral
Program in Biology, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki 305-8572, Japan
| | - Rikuri Morita
- Center
for Computational Sciences, University of
Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Yasuteru Shigeta
- Center
for Computational Sciences, University of
Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Ryuhei Harada
- Center
for Computational Sciences, University of
Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| |
Collapse
|
31
|
Xu K, Zhong J, Li J, Cao Y, Wei L. Structure features of Streptococcus pneumoniae FabG and virtual screening of allosteric inhibitors. Front Mol Biosci 2024; 11:1472252. [PMID: 39398278 PMCID: PMC11467476 DOI: 10.3389/fmolb.2024.1472252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
Streptococcus pneumoniae, a gram-positive bacterium, is responsible for diverse infections globally, and its antibiotic resistance presents significant challenges to medical advancements. It is imperative to employ various strategies to identify antibiotics. 3-oxoacyl-[acyl-carrier-protein] reductase (FabG) is a key component in the type II fatty acid synthase (FAS II) system, which is a developing target for new anti-streptococcal drugs. We first demonstrated the function of SpFabG in vivo and in vitro and the 2 Å SpFabG structure was elucidated using X-ray diffraction technique. It was observed that the NADPH binding promotes the transformation from tetramers to dimers in solution, suggesting dimers but not tetramer may be the active conformation. By comparing the structures of FabG homologues, we have identified the conserved tetramerization site and further confirmed the mechanism that the tetramerization site mutation leads to a loss of function and destabilization through mutagenesis experiments. Starting from 533,600 compounds, we proceeded with a sequential workflow involving pharmacophore-based virtual screening, molecular docking, and binding energy calculations. Combining all the structural analysis, we identified L1, L2 and L5 as a promising candidate for SpFabG inhibitor, based on the most stable binding mode in comparison to other evaluated inhibitors.
Collapse
Affiliation(s)
- Kaimin Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jianliang Zhong
- Molecular Cancer Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Jing Li
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Yulu Cao
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Lai Wei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Ophthalmology, The First Affiliated Hospital, University of South China, Hengyang, China
| |
Collapse
|
32
|
de Jager M, Kolbeck PJ, Vanderlinden W, Lipfert J, Filion L. Exploring protein-mediated compaction of DNA by coarse-grained simulations and unsupervised learning. Biophys J 2024; 123:3231-3241. [PMID: 39044429 PMCID: PMC11427786 DOI: 10.1016/j.bpj.2024.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/18/2024] [Accepted: 07/18/2024] [Indexed: 07/25/2024] Open
Abstract
Protein-DNA interactions and protein-mediated DNA compaction play key roles in a range of biological processes. The length scales typically involved in DNA bending, bridging, looping, and compaction (≥1 kbp) are challenging to address experimentally or by all-atom molecular dynamics simulations, making coarse-grained simulations a natural approach. Here, we present a simple and generic coarse-grained model for DNA-protein and protein-protein interactions and investigate the role of the latter in the protein-induced compaction of DNA. Our approach models the DNA as a discrete worm-like chain. The proteins are treated in the grand canonical ensemble, and the protein-DNA binding strength is taken from experimental measurements. Protein-DNA interactions are modeled as an isotropic binding potential with an imposed binding valency without specific assumptions about the binding geometry. To systematically and quantitatively classify DNA-protein complexes, we present an unsupervised machine learning pipeline that receives a large set of structural order parameters as input, reduces the dimensionality via principal-component analysis, and groups the results using a Gaussian mixture model. We apply our method to recent data on the compaction of viral genome-length DNA by HIV integrase and find that protein-protein interactions are critical to the formation of looped intermediate structures seen experimentally. Our methodology is broadly applicable to DNA-binding proteins and protein-induced DNA compaction and provides a systematic and semi-quantitative approach for analyzing their mesoscale complexes.
Collapse
Affiliation(s)
- Marjolein de Jager
- Soft Condensed Matter and Biophysics, Debye Institute for Nanomaterials Science, Utrecht University, Utrecht, the Netherlands.
| | - Pauline J Kolbeck
- Soft Condensed Matter and Biophysics, Debye Institute for Nanomaterials Science, Utrecht University, Utrecht, the Netherlands; Department of Physics and Center for NanoScience, LMU, Munich, Germany
| | - Willem Vanderlinden
- Soft Condensed Matter and Biophysics, Debye Institute for Nanomaterials Science, Utrecht University, Utrecht, the Netherlands; Department of Physics and Center for NanoScience, LMU, Munich, Germany; School of Physics and Astronomy, University of Edinburgh, Scotland, United Kingdom
| | - Jan Lipfert
- Soft Condensed Matter and Biophysics, Debye Institute for Nanomaterials Science, Utrecht University, Utrecht, the Netherlands; Department of Physics and Center for NanoScience, LMU, Munich, Germany
| | - Laura Filion
- Soft Condensed Matter and Biophysics, Debye Institute for Nanomaterials Science, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
33
|
Ye L, Ajuyo NMC, Wu Z, Yuan N, Xiao Z, Gu W, Zhao J, Pei Y, Min Y, Wang D. Molecular Integrative Study on Inhibitory Effects of Pentapeptides on Polymerization and Cell Toxicity of Amyloid-β Peptide (1-42). Curr Issues Mol Biol 2024; 46:10160-10179. [PMID: 39329958 PMCID: PMC11431437 DOI: 10.3390/cimb46090606] [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: 07/30/2024] [Revised: 09/07/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024] Open
Abstract
Alzheimer's Disease (AD) is a multifaceted neurodegenerative disease predominantly defined by the extracellular accumulation of amyloid-β (Aβ) peptide. In light of this, in the past decade, several clinical approaches have been used aiming at developing peptides for therapeutic use in AD. The use of cationic arginine-rich peptides (CARPs) in targeting protein aggregations has been on the rise. Also, the process of peptide development employing computational approaches has attracted a lot of attention recently. Using a structure database containing pentapeptides made from 20 L-α amino acids, we employed molecular docking to sort pentapeptides that can bind to Aβ42, then performed molecular dynamics (MD) analyses, including analysis of the binding stability, interaction energy, and binding free energy to screen ligands. Transmission electron microscopy (TEM), circular dichroism (CD), thioflavin T (ThT) fluorescence detection of Aβ42 polymerization, MTT (3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide) assay, and the flow cytometry of reactive oxygen species (ROS) were carried out to evaluate the influence of pentapeptides on the aggregation and cell toxicity of Aβ42. Two pentapeptides (TRRRR and ARRGR) were found to have strong effects on inhibiting the aggregation of Aβ42 and reducing the toxicity of Aβ42 secreted by SH-SY5Y cells, including cell death, reactive oxygen species (ROS) production, and apoptosis.
Collapse
Affiliation(s)
- Lianmeng Ye
- Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Nuela Manka'a Che Ajuyo
- Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
| | - Zhongyun Wu
- Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Nan Yuan
- Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Zhengpan Xiao
- Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Wenyu Gu
- Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Jiazheng Zhao
- Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Yechun Pei
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Yi Min
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
- Department of Biotechnology, School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Dayong Wang
- Key Laboratory of Tropical Biological Resources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, One Health Cooperative Innovation Center, Hainan University, Haikou 570228, China
| |
Collapse
|
34
|
Raghuraman P, Park S. Molecular simulation reveals that pathogenic mutations in BTB/ANK domains of Arabidopsis thaliana NPR1 circumscribe the EDS1-mediated immune regulation. JOURNAL OF PLANT PHYSIOLOGY 2024; 303:154345. [PMID: 39353309 DOI: 10.1016/j.jplph.2024.154345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 10/04/2024]
Abstract
The NPR1 (nonexpressor of pathogenesis-related genes 1) is a key regulator of the salicylic-acid-mediated immune response caused by pathogens in Arabidopsis thaliana. Mutations C150Y and H334Y in the BTB/ANK domains of NPR1 inhibit the defense response, and transcriptional co-activity with enhanced disease susceptibility 1 (EDS1) has been revealed experimentally. This study examined the conformational changes and reduced NPR1-EDS1 interaction upon mutation using a molecular dynamics simulation. Initially, BTBC150YNPR1 and ANKH334YNPR1 were categorized as pathological mutations rather than others based on sequence conservation. A distant ortholog was used to map the common residues shared among the wild-type because the mutations were highly conserved. Overall, 179 of 373 residues were determining the secondary structures and fold versatility of conformations. In addition, the mutational hotspots Cys150, Asp152, Glu153, Cys155, His157, Cys160, His334, Arg339 and Lys370 were crucial for oligomer-to-monomer exchange. Subsequently, the atomistic simulations with free energy (MM/PB(GB)SA) calculations predicted structural displacements engaging in the N-termini α5133-178α7 linker connecting the central ANK regions (α13260-290α14 and α18320-390α22), where prominent long helices (α516) and short helices (α310) replaced with β-turns and loops disrupting hydrogen bonds and salt bridges in both mutants implicating functional regulation and activation. Furthermore, the mutation repositions the intact stability of multiple regions (L13C149-N356α20BTB/ANK-α17W301-E357α21N-ter/coiled-coil) compromising a dynamic interaction of NPR1-EDS1. By unveiling the transitions between the distinct functions of mutational perception, this study paves the way for future investigation to orchestrate additive host-adapted transcriptional reprogramming that controls defense-related regulatory mechanisms of NPR1s in plants.
Collapse
Affiliation(s)
- P Raghuraman
- Department of Life Sciences, Yeungnam University, Gyeongsan, Gyeongsangbuk-do, 38541, Republic of Korea
| | - SeonJoo Park
- Department of Life Sciences, Yeungnam University, Gyeongsan, Gyeongsangbuk-do, 38541, Republic of Korea.
| |
Collapse
|
35
|
Neubergerová M, Pleskot R. Plant protein-lipid interfaces studied by molecular dynamics simulations. JOURNAL OF EXPERIMENTAL BOTANY 2024; 75:5237-5250. [PMID: 38761107 DOI: 10.1093/jxb/erae228] [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: 12/14/2023] [Accepted: 05/16/2024] [Indexed: 05/20/2024]
Abstract
The delineation of protein-lipid interfaces is essential for understanding the mechanisms of various membrane-associated processes crucial to plant development and growth, including signalling, trafficking, and membrane transport. Due to their highly dynamic nature, the precise characterization of lipid-protein interactions by experimental techniques is challenging. Molecular dynamics simulations provide a powerful computational alternative with a spatial-temporal resolution allowing the atomistic-level description. In this review, we aim to introduce plant scientists to molecular dynamics simulations. We describe different steps of performing molecular dynamics simulations and provide a broad survey of molecular dynamics studies investigating plant protein-lipid interfaces. Our aim is also to illustrate that combining molecular dynamics simulations with artificial intelligence-based protein structure determination opens up unprecedented possibilities for future investigations of dynamic plant protein-lipid interfaces.
Collapse
Affiliation(s)
- Michaela Neubergerová
- Institute of Experimental Botany, Czech Academy of Sciences, Prague, Czech Republic
- Department of Experimental Plant Biology, Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Roman Pleskot
- Institute of Experimental Botany, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
36
|
Nencini R, Tempra C, Biriukov D, Riopedre-Fernandez M, Cruces Chamorro V, Polák J, Mason PE, Ondo D, Heyda J, Ollila OHS, Jungwirth P, Javanainen M, Martinez-Seara H. Effective Inclusion of Electronic Polarization Improves the Description of Electrostatic Interactions: The prosECCo75 Biomolecular Force Field. J Chem Theory Comput 2024; 20:7546-7559. [PMID: 39186899 PMCID: PMC11391585 DOI: 10.1021/acs.jctc.4c00743] [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: 08/28/2024]
Abstract
prosECCo75 is an optimized force field effectively incorporating electronic polarization via charge scaling. It aims to enhance the accuracy of nominally nonpolarizable molecular dynamics simulations for interactions in biologically relevant systems involving water, ions, proteins, lipids, and saccharides. Recognizing the inherent limitations of nonpolarizable force fields in precisely modeling electrostatic interactions essential for various biological processes, we mitigate these shortcomings by accounting for electronic polarizability in a physically rigorous mean-field way that does not add to computational costs. With this scaling of (both integer and partial) charges within the CHARMM36 framework, prosECCo75 addresses overbinding artifacts. This improves agreement with experimental ion binding data across a broad spectrum of systems─lipid membranes, proteins (including peptides and amino acids), and saccharides─without compromising their biomolecular structures. prosECCo75 thus emerges as a computationally efficient tool providing enhanced accuracy and broader applicability in simulating the complex interplay of interactions between ions and biomolecules, pivotal for improving our understanding of many biological processes.
Collapse
Affiliation(s)
- Ricky Nencini
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, CZ-160 00 Prague 6, Czech Republic
- Institute of Biotechnology, University of Helsinki, Viikinkaari 5, FI-00790 Helsinki, Finland
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5, FI-00790 Helsinki, Finland
| | - Carmelo Tempra
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, CZ-160 00 Prague 6, Czech Republic
| | - Denys Biriukov
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, CZ-160 00 Prague 6, Czech Republic
- CEITEC─Central European Institute of Technology, Masaryk University, Kamenice 753/5, CZ-62500 Brno, Czech Republic
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 753/5, CZ-62500 Brno, Czech Republic
| | - Miguel Riopedre-Fernandez
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, CZ-160 00 Prague 6, Czech Republic
| | - Victor Cruces Chamorro
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, CZ-160 00 Prague 6, Czech Republic
| | - Jakub Polák
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Technická 5, CZ-166 28 Prague 6, Czech Republic
| | - Philip E Mason
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, CZ-160 00 Prague 6, Czech Republic
| | - Daniel Ondo
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Technická 5, CZ-166 28 Prague 6, Czech Republic
| | - Jan Heyda
- Department of Physical Chemistry, University of Chemistry and Technology, Prague, Technická 5, CZ-166 28 Prague 6, Czech Republic
| | - O H Samuli Ollila
- Institute of Biotechnology, University of Helsinki, Viikinkaari 5, FI-00790 Helsinki, Finland
- VTT Technical Research Centre of Finland, Tietotie 2, FI-02150 Espoo, Finland
| | - Pavel Jungwirth
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, CZ-160 00 Prague 6, Czech Republic
| | - Matti Javanainen
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, CZ-160 00 Prague 6, Czech Republic
- Institute of Biotechnology, University of Helsinki, Viikinkaari 5, FI-00790 Helsinki, Finland
| | - Hector Martinez-Seara
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nám. 2, CZ-160 00 Prague 6, Czech Republic
| |
Collapse
|
37
|
Son A, Kim W, Park J, Lee W, Lee Y, Choi S, Kim H. Utilizing Molecular Dynamics Simulations, Machine Learning, Cryo-EM, and NMR Spectroscopy to Predict and Validate Protein Dynamics. Int J Mol Sci 2024; 25:9725. [PMID: 39273672 PMCID: PMC11395565 DOI: 10.3390/ijms25179725] [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: 08/01/2024] [Revised: 09/06/2024] [Accepted: 09/07/2024] [Indexed: 09/15/2024] Open
Abstract
Protein dynamics play a crucial role in biological function, encompassing motions ranging from atomic vibrations to large-scale conformational changes. Recent advancements in experimental techniques, computational methods, and artificial intelligence have revolutionized our understanding of protein dynamics. Nuclear magnetic resonance spectroscopy provides atomic-resolution insights, while molecular dynamics simulations offer detailed trajectories of protein motions. Computational methods applied to X-ray crystallography and cryo-electron microscopy (cryo-EM) have enabled the exploration of protein dynamics, capturing conformational ensembles that were previously unattainable. The integration of machine learning, exemplified by AlphaFold2, has accelerated structure prediction and dynamics analysis. These approaches have revealed the importance of protein dynamics in allosteric regulation, enzyme catalysis, and intrinsically disordered proteins. The shift towards ensemble representations of protein structures and the application of single-molecule techniques have further enhanced our ability to capture the dynamic nature of proteins. Understanding protein dynamics is essential for elucidating biological mechanisms, designing drugs, and developing novel biocatalysts, marking a significant paradigm shift in structural biology and drug discovery.
Collapse
Affiliation(s)
- Ahrum Son
- Department of Molecular Medicine, Scripps Research, San Diego, CA 92037, USA
| | - Woojin Kim
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Jongham Park
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Wonseok Lee
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Yerim Lee
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Seongyun Choi
- Department of Convergent Bioscience and Informatics, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Hyunsoo Kim
- Department of Bio-AI Convergence, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
- Department of Convergent Bioscience and Informatics, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
- Protein AI Design Institute, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
- SCICS, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| |
Collapse
|
38
|
Lowry GV, Giraldo JP, Steinmetz NF, Avellan A, Demirer GS, Ristroph KD, Wang GJ, Hendren CO, Alabi CA, Caparco A, da Silva W, González-Gamboa I, Grieger KD, Jeon SJ, Khodakovskaya MV, Kohay H, Kumar V, Muthuramalingam R, Poffenbarger H, Santra S, Tilton RD, White JC. Towards realizing nano-enabled precision delivery in plants. NATURE NANOTECHNOLOGY 2024; 19:1255-1269. [PMID: 38844663 DOI: 10.1038/s41565-024-01667-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 03/27/2024] [Indexed: 09/18/2024]
Abstract
Nanocarriers (NCs) that can precisely deliver active agents, nutrients and genetic materials into plants will make crop agriculture more resilient to climate change and sustainable. As a research field, nano-agriculture is still developing, with significant scientific and societal barriers to overcome. In this Review, we argue that lessons can be learned from mammalian nanomedicine. In particular, it may be possible to enhance efficiency and efficacy by improving our understanding of how NC properties affect their interactions with plant surfaces and biomolecules, and their ability to carry and deliver cargo to specific locations. New tools are required to rapidly assess NC-plant interactions and to explore and verify the range of viable targeting approaches in plants. Elucidating these interactions can lead to the creation of computer-generated in silico models (digital twins) to predict the impact of different NC and plant properties, biological responses, and environmental conditions on the efficiency and efficacy of nanotechnology approaches. Finally, we highlight the need for nano-agriculture researchers and social scientists to converge in order to develop sustainable, safe and socially acceptable NCs.
Collapse
Affiliation(s)
- Gregory V Lowry
- Civil and Environmental Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
| | - Juan Pablo Giraldo
- Botany and Plant Sciences, University of California, Riverside, Riverside, CA, USA.
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California San Diego, San Diego, CA, USA
- Department of Bioengineering, University of California San Diego, San Diego, CA, USA
- Department of Radiology, University of California San Diego, San Diego, CA, USA
- Center for Nano-ImmunoEngineering, University of California San Diego, San Diego, CA, USA
- Shu and K.C. Chien and Peter Farrell Collaboratory, University of California San Diego, San Diego, CA, USA
- Center for Engineering in Cancer, Institute of Engineering in Medicine, University of California San Diego, San Diego, CA, USA
- Moores Cancer Center, University of California, University of California San Diego, San Diego, CA, USA
- Institute for Materials Discovery and Design, University of California San Diego, San Diego, CA, USA
| | | | - Gozde S Demirer
- Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Kurt D Ristroph
- Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, USA
| | - Gerald J Wang
- Civil and Environmental Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Christine O Hendren
- Geological and Environmental Sciences, Appalachian State University, Boone, NC, USA
| | | | - Adam Caparco
- Department of NanoEngineering, University of California San Diego, San Diego, CA, USA
| | | | | | - Khara D Grieger
- Applied Ecology, North Carolina State University, Raleigh, NC, USA
| | - Su-Ji Jeon
- Botany and Plant Sciences, University of California, Riverside, Riverside, CA, USA
| | | | - Hagay Kohay
- Civil and Environmental Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Vivek Kumar
- Civil and Environmental Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | | | | | - Swadeshmukul Santra
- Department of Chemistry and Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Robert D Tilton
- Chemical Engineering and Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Jason C White
- The Connecticut Agricultural Research Station, New Haven, CT, USA
| |
Collapse
|
39
|
Huang Y, Zhang H, Lin Z, Wei Y, Xi W. RevGraphVAMP: A protein molecular simulation analysis model combining graph convolutional neural networks and physical constraints. Methods 2024; 229:163-174. [PMID: 38972499 DOI: 10.1016/j.ymeth.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/09/2024] Open
Abstract
Molecular dynamics simulation is a crucial research domain within the life sciences, focusing on comprehending the mechanisms of biomolecular interactions at atomic scales. Protein simulation, as a critical subfield, often utilizes MD for implementation, with trajectory data play a pivotal role in drug discovery. The advancement of high-performance computing and deep learning technology becomes popular and critical to predict protein properties from vast trajectory data, posing challenges regarding data features extraction from the complicated simulation data and dimensionality reduction. Simultaneously, it is essential to provide a meaningful explanation of the biological mechanism behind dimensionality. To tackle this challenge, we propose a new unsupervised model named RevGraphVAMP to intelligently analyze the simulation trajectory. This model is based on the variational approach for Markov processes (VAMP) and integrates graph convolutional neural networks and physical constraint optimization to enhance the learning performance. Additionally, we introduce attention mechanism to assess the importance of key interaction region, facilitating the interpretation of molecular mechanism. In comparison to other VAMPNets models, our model showcases competitive performance, improved accuracy in state transition prediction, as demonstrated through its application to two public datasets and the Shank3-Rap1 complex, which is associated with autism spectrum disorder. Moreover, it enhanced dimensionality reduction discrimination across different substates and provides interpretable results for protein structural characterization.
Collapse
Affiliation(s)
- Ying Huang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Huiling Zhang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; College of Mathematics and Informatics, South China Agricultural University, Guangzhou, 510642, China
| | - Zhenli Lin
- Department of Ophthalmology, Shenzhen University General Hospital, Shenzhen 518055, China
| | - Yanjie Wei
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen 518107, China.
| | - Wenhui Xi
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen 518107, China.
| |
Collapse
|
40
|
Raza S, Sarkar D, Chan LJG, Mae J, Sutter M, Petzold CJ, Kerfeld CA, Ralston CY, Gupta S, Vermaas JV. Comparative Pore Structure and Dynamics for Bacterial Microcompartment Shell Protein Assemblies in Sheets or Shells. ACS OMEGA 2024; 9:35503-35514. [PMID: 39184480 PMCID: PMC11339822 DOI: 10.1021/acsomega.4c02406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 08/27/2024]
Abstract
Bacterial microcompartments (BMCs) are protein-bound organelles found in some bacteria that encapsulate enzymes for enhanced catalytic activity. These compartments spatially sequester enzymes within semipermeable shell proteins, analogous to many membrane-bound organelles. The shell proteins assemble into multimeric tiles; hexamers, trimers, and pentamers, and these tiles self-assemble into larger assemblies with icosahedral symmetry. While icosahedral shells are the predominant form in vivo, the tiles can also form nanoscale cylinders or sheets. The individual multimeric tiles feature central pores that are key to regulating transport across the protein shell. Our primary interest is to quantify pore shape changes in response to alternative component morphologies at the nanoscale. We used molecular modeling tools to develop atomically detailed models for both planar sheets of tiles and curved structures representative of the complete shells found in vivo. Subsequently, these models were animated using classical molecular dynamics simulations. From the resulting trajectories, we analyzed the overall structural stability, water accessibility to individual residues, water residence time, and pore geometry for the hexameric and trimeric protein tiles from the Haliangium ochraceum model BMC shell. These exhaustive analyses suggest no substantial variation in pore structure or solvent accessibility between the flat and curved shell geometries. We additionally compare our analysis to hydroxyl radical footprinting data to serve as a check against our simulation results, highlighting specific residues where water molecules are bound for a long time. Although with little variation in morphology or water interaction, we propose that the planar and capsular morphology can be used interchangeably when studying permeability through BMC pores.
Collapse
Affiliation(s)
- Saad Raza
- MSU-DOE
Plant Research Laboratory, Michigan State
University, East Lansing, Michigan 48824, United States
| | - Daipayan Sarkar
- MSU-DOE
Plant Research Laboratory, Michigan State
University, East Lansing, Michigan 48824, United States
| | - Leanne Jade G. Chan
- Biological
Systems and Engineering Division, Lawrence
Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Joshua Mae
- MSU-DOE
Plant Research Laboratory, Michigan State
University, East Lansing, Michigan 48824, United States
| | - Markus Sutter
- MSU-DOE
Plant Research Laboratory, Michigan State
University, East Lansing, Michigan 48824, United States
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Christopher J. Petzold
- Biological
Systems and Engineering Division, Lawrence
Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Cheryl A. Kerfeld
- MSU-DOE
Plant Research Laboratory, Michigan State
University, East Lansing, Michigan 48824, United States
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
- Department
of Biochemistry and Molecular Biology, Michigan
State University, East Lansing, Michigan 48824, United States
| | - Corie Y. Ralston
- Molecular
Foundry Division, Lawrence Berkeley National
Laboratory, Berkeley, California 94720, United States
| | - Sayan Gupta
- Molecular
Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Josh V. Vermaas
- MSU-DOE
Plant Research Laboratory, Michigan State
University, East Lansing, Michigan 48824, United States
- Department
of Biochemistry and Molecular Biology, Michigan
State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
41
|
Zhong L, Wang Z, Ye X, Cui J, Wang Z, Jia S. Molecular simulations guide immobilization of lipase on nest-like ZIFs with regulatable hydrophilic/hydrophobic surface. J Colloid Interface Sci 2024; 667:199-211. [PMID: 38636222 DOI: 10.1016/j.jcis.2024.04.075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/24/2024] [Accepted: 04/12/2024] [Indexed: 04/20/2024]
Abstract
The catalytic performance of immobilized lipase is greatly influenced by functional support, which attracts growing interest for designing supports to achieve their promotive catalytic activity. Many lipases bind strongly to hydrophobic surfaces where they undergo interfacial activation. Herein, the behavioral differences of lipases with distinct lid structures on interfaces of varying hydrophobicity levels were firstly investigated by molecular simulations. It was found that a reasonable hydrophilic/hydrophobic surface could facilitate the lipase to undergo interfacial activation. Building on these findings, a novel "nest"-like superhydrophobic ZIFs (ZIFN) composed of hydrophobic ligands was prepared for the first time and used to immobilize lipase from Aspergillus oryzae (AOL@ZIFN). The AOL@ZIFN exhibited 2.0-folds higher activity than free lipase in the hydrolysis of p-Nitrophenyl palmitate (p-NPP). Especially, the modification of superhydrophobic ZIFN with an appropriate amount of hydrophilic tannic acid can significantly improve the activity of the immobilized lipase (AOL@ZIFN-TA). The AOL@ZIFN-TA exhibited 30-folds higher activity than free lipase, and still maintained 82% of its initial activity after 5 consecutive cycles, indicating good reusability. These results demonstrated that nanomaterials with rational arrangement of the hydrophilic/hydrophobic surface could facilitate the lipase to undergo interfacial activation and improve its activity, displaying the potential of the extensive application.
Collapse
Affiliation(s)
- Le Zhong
- State Key Laboratory of Food Nutrition and Safety, Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, No. 29, 13th Avenue, Tianjin Economic and Technological Development Area (TEDA), Tianjin 300457, PR China
| | - Zhongjie Wang
- State Key Laboratory of Food Nutrition and Safety, Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, No. 29, 13th Avenue, Tianjin Economic and Technological Development Area (TEDA), Tianjin 300457, PR China
| | - Xiaohong Ye
- State Key Laboratory of Food Nutrition and Safety, Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, No. 29, 13th Avenue, Tianjin Economic and Technological Development Area (TEDA), Tianjin 300457, PR China
| | - Jiandong Cui
- State Key Laboratory of Food Nutrition and Safety, Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, No. 29, 13th Avenue, Tianjin Economic and Technological Development Area (TEDA), Tianjin 300457, PR China.
| | - Ziyuan Wang
- State Key Laboratory of Food Nutrition and Safety, Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, No. 29, 13th Avenue, Tianjin Economic and Technological Development Area (TEDA), Tianjin 300457, PR China.
| | - Shiru Jia
- State Key Laboratory of Food Nutrition and Safety, Laboratory of Industrial Fermentation Microbiology, Ministry of Education, Tianjin University of Science and Technology, No. 29, 13th Avenue, Tianjin Economic and Technological Development Area (TEDA), Tianjin 300457, PR China
| |
Collapse
|
42
|
Iñaki Gamero-Barraza J, Antonio Pámanes-Carrasco G, Delgado E, Patricia Cabrales-Arellano C, Medrano-Roldán H, Gallegos-Ibáñez D, Wedwitschka H, Reyes-Jáquez D. Computational modelling of extrusion process temperatures on the interactions between black soldier fly larvae protein and corn flour starch. FOOD CHEMISTRY. MOLECULAR SCIENCES 2024; 8:100202. [PMID: 38586156 PMCID: PMC10995973 DOI: 10.1016/j.fochms.2024.100202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/09/2024]
Abstract
Insects such as the black soldier fly (BSF) are recently being studied as food sources to address concerns about how to meet the food demand of the growing world population, as conventional production lines for meat proteins are currently unsustainable sources. Studies have been conducted evaluating the use of insect proteins to produce extruded foods such as expanded snacks and meat analogues. However, this field of study is still quite new and not much has been studied beyond digestibility and growth performance. The purpose of this work was to evaluate the compatibility of protein extracted from BSF flour with corn flour starch within an extruded balanced shrimp feed model through molecular dynamics simulations, for which cohesive energy density and solubility parameter (δ) of both components were determined. The calculations' results for the protein molecule systems yielded an average δ of 14.961 MPa0.5, while the δ for starch was calculated to be 23.166 MPa0.5. The range of difference between both δ (10 > δ > 7) suggests that the interaction of the BSF protein with corn starch is of a semi-miscible nature. These results suggest that it is possible to obtain a stable starch-protein mixture through the extrusion process.
Collapse
Affiliation(s)
- Jorge Iñaki Gamero-Barraza
- TecNM/I.T.Durango. Posgrado en Ingeniería Bioquímica Felipe Pescador 1803, Nueva Vizcaya, 34080 Durango, Dgo., Mexico
| | - Gerardo Antonio Pámanes-Carrasco
- Facultad de Medicina Veterinaria y Zootecnia de la Universidad Juárez del Estado Durango/ Durango - Mezquital Km 11.5, 34307 Durango, Dgo., Mexico
| | - Efrén Delgado
- Food Science and Technology, Department of Family and Consumer Sciences, New Mexico State University, P.O. Box 30001, Las Cruces, NM 88003-8001, USA
| | | | - Hiram Medrano-Roldán
- TecNM/I.T.Durango. Posgrado en Ingeniería Bioquímica Felipe Pescador 1803, Nueva Vizcaya, 34080 Durango, Dgo., Mexico
| | - Daniela Gallegos-Ibáñez
- Department of Biochemical Conversion, Deutsches Biomasseforschungszentrum gemeinnützige GmbH, Torgauer Straße116, 04347, Leipzig, Germany
| | - Harald Wedwitschka
- Department of Biochemical Conversion, Deutsches Biomasseforschungszentrum gemeinnützige GmbH, Torgauer Straße116, 04347, Leipzig, Germany
| | - Damián Reyes-Jáquez
- TecNM/I.T.Durango. Posgrado en Ingeniería Bioquímica Felipe Pescador 1803, Nueva Vizcaya, 34080 Durango, Dgo., Mexico
| |
Collapse
|
43
|
Akki AJ, Patil SA, Hungund S, Sahana R, Patil MM, Kulkarni RV, Raghava Reddy K, Zameer F, Raghu AV. Advances in Parkinson's disease research - A computational network pharmacological approach. Int Immunopharmacol 2024; 139:112758. [PMID: 39067399 DOI: 10.1016/j.intimp.2024.112758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Parkinson's disease (PD), the second most prevalent neurodegenerative disorder, is projected to see a significant rise in incidence over the next three decades. The precise treatment of PD remains a formidable challenge, prompting ongoing research into early diagnostic methodologies. Network pharmacology, a burgeoning field grounded in systems biology, examines the intricate networks of biological systems to identify critical signal nodes, facilitating the development of multi-target therapeutic molecules. This approach systematically maps the components of Parkinson's disease, thereby reducing its complexity. In this review, we explore the application of network pharmacology workflows in PD, discuss the techniques employed in this field, and evaluate the current advancements and status of network pharmacology in the context of Parkinson's disease. The comprehensive insights will pave newer paths to explore early disease biomarkers and to develop diagnosis with a holistic in silico, in vitro, in vivo and clinical studies.
Collapse
Affiliation(s)
- Ali Jawad Akki
- Faculty of Science and Technology, BLDE (Deemed-to-be University), Vijayapura 586 103, India
| | - Shruti A Patil
- Faculty of Science and Technology, BLDE (Deemed-to-be University), Vijayapura 586 103, India
| | - Sphoorty Hungund
- Faculty of Science and Technology, BLDE (Deemed-to-be University), Vijayapura 586 103, India
| | - R Sahana
- Department of Computer Science and Engineering, RV Institute of Technology and Management, 560 076 Bengaluru, India
| | - Malini M Patil
- Department of Computer Science and Engineering, RV Institute of Technology and Management, 560 076 Bengaluru, India.
| | - Raghavendra V Kulkarni
- Faculty of Science and Technology, BLDE (Deemed-to-be University), Vijayapura 586 103, India
| | - K Raghava Reddy
- School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, NSW 12 2006, Australia
| | - Farhan Zameer
- Department of Dravyaguna (Ayurveda Pharmacology), Alva's Ayurveda Medical College, and PathoGutOmics Laboratory, ATMA Research Centre, Dakshina Kannada 574 227, India.
| | - Anjanapura V Raghu
- Department of Basic Sciences, Faculty of Engineering and Technology, CMR University, 562149 Bangalore, India.
| |
Collapse
|
44
|
Mondal S, Sauer MA, Heyden M. Exploring Conformational Landscapes Along Anharmonic Low-Frequency Vibrations. J Phys Chem B 2024; 128:7112-7120. [PMID: 38986052 PMCID: PMC11584282 DOI: 10.1021/acs.jpcb.4c02743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
We aim to automatize the identification of collective variables to simplify and speed up enhanced sampling simulations of conformational dynamics in biomolecules. We focus on anharmonic low-frequency vibrations that exhibit fluctuations on time scales faster than conformational transitions but describe a path of least resistance toward structural change. A key challenge is that harmonic approximations are ill-suited to characterize these vibrations, which are observed at far-infrared frequencies and are easily excited by thermal collisions at room temperature. Here, we approached this problem with a frequency-selective anharmonic (FRESEAN) mode analysis that does not rely on harmonic approximations and successfully isolates anharmonic low-frequency vibrations from short molecular dynamics simulation trajectories. We applied FRESEAN mode analysis to simulations of alanine dipeptide, a common test system for enhanced sampling simulation protocols, and compared the performance of isolated low-frequency vibrations to conventional user-defined collective variables (here backbone dihedral angles) in enhanced sampling simulations. The comparison shows that enhanced sampling along anharmonic low-frequency vibrations not only reproduces known conformational dynamics but can even further improve the sampling of slow transitions compared to user-defined collective variables. Notably, free energy surfaces spanned by low-frequency anharmonic vibrational modes exhibit lower barriers associated with conformational transitions relative to representations in backbone dihedral space. We thus conclude that anharmonic low-frequency vibrations provide a promising path for highly effective and fully automated enhanced sampling simulations of conformational dynamics in biomolecules.
Collapse
Affiliation(s)
- Souvik Mondal
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Michael A Sauer
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| | - Matthias Heyden
- School of Molecular Sciences, Arizona State University, Tempe, Arizona 85287, United States
| |
Collapse
|
45
|
Ansell TB, Healy M, Coupland CE, Sansom MSP, Siebold C. Mapping structural and dynamic divergence across the MBOAT family. Structure 2024; 32:1011-1022.e3. [PMID: 38636523 DOI: 10.1016/j.str.2024.03.014] [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: 12/14/2023] [Revised: 02/09/2024] [Accepted: 03/22/2024] [Indexed: 04/20/2024]
Abstract
Membrane-bound O-acyltransferases (MBOATs) are membrane-embedded enzymes that catalyze acyl chain transfer to a diverse group of substrates, including lipids, small molecules, and proteins. MBOATs share a conserved structural core, despite wide-ranging functional specificity across both prokaryotes and eukaryotes. The structural basis of catalytic specificity, regulation and interactions with the surrounding environment remain uncertain. Here, we combine comparative molecular dynamics (MD) simulations with bioinformatics to assess molecular and interactional divergence across the family. In simulations, MBOATs differentially distort the bilayer depending on their substrate type. Additionally, we identify lipid binding sites surrounding reactant gates in the surrounding membrane. Complementary bioinformatic analyses reveal a conserved role for re-entrant loop-2 in MBOAT fold stabilization and a key hydrogen bond bridging DGAT1 dimerization. Finally, we predict differences in MBOAT solvation and water gating properties. These data are pertinent to the design of MBOAT-specific inhibitors that encompass dynamic information within cellular mimetic environments.
Collapse
Affiliation(s)
- T Bertie Ansell
- Department of Biochemistry, South Parks Road, Oxford OX1 3QU, UK; Division of CryoEM and Bioimaging, SSRL, SLAC National Accelerator Laboratory, Menlo Park, CA 94025, USA; Department of Biology, Stanford University, Stanford, CA 94305, USA.
| | - Megan Healy
- Department of Biochemistry, South Parks Road, Oxford OX1 3QU, UK
| | - Claire E Coupland
- Division of Structural Biology, Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK; Molecular Medicine Program, The Hospital for Sick Children, 686 Bay Street, Toronto M5G 0A4, Canada
| | - Mark S P Sansom
- Department of Biochemistry, South Parks Road, Oxford OX1 3QU, UK
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, Roosevelt Drive, Oxford OX3 7BN, UK
| |
Collapse
|
46
|
Zeng Q, Hu H, Huang Z, Guo A, Lu S, Tong W, Zhang Z, Shen T. Active and machine learning-enhanced discovery of new FGFR3 inhibitor, Rhapontin, through virtual screening of receptor structures and anti-cancer activity assessment. Front Mol Biosci 2024; 11:1413214. [PMID: 38919748 PMCID: PMC11196408 DOI: 10.3389/fmolb.2024.1413214] [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: 04/19/2024] [Accepted: 05/23/2024] [Indexed: 06/27/2024] Open
Abstract
Introduction: This study bridges traditional remedies and modern pharmacology by exploring the synergy between natural compounds and Ceritinib in treating Non-Small Cell Lung Cancer (NSCLC), aiming to enhance efficacy and reduce toxicities. Methods: Using a combined approach of computational analysis, machine learning, and experimental procedures, we identified and analyzed PD173074, Isoquercitrin, and Rhapontin as potential inhibitors of fibroblast growth factor receptor 3 (FGFR3). Machine learning algorithms guided the initial selection, followed by Quantitative Structure-Activity Relationship (QSAR) modeling and molecular dynamics simulations to evaluate the interaction dynamics and stability of Rhapontin. Physicochemical assessments further verified its drug-like properties and specificity. Results: Our experiments demonstrate that Rhapontin, when combined with Ceritinib, significantly suppresses tumor activity in NSCLC while sparing healthy cells. The molecular simulations and physicochemical evaluations confirm Rhapontin's stability and favorable interaction with FGFR3, highlighting its potential as an effective adjunct in NSCLC therapy. Discussion: The integration of natural compounds with established cancer therapies offers a promising avenue for enhancing treatment outcomes in NSCLC. By combining the ancient wisdom of natural remedies with the precision of modern science, this study contributes to evolving cancer treatment paradigms, potentially mitigating the side effects associated with current therapies.
Collapse
Affiliation(s)
- Qingxin Zeng
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haichuan Hu
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengwei Huang
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Aotian Guo
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sheng Lu
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenbin Tong
- Department of Thoracic Surgery, Longyou County People’s Hospital, Hangzhou, China
| | - Zhongheng Zhang
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Hangzhou, China
| | - Tao Shen
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
47
|
Ghosh D, Biswas A, Radhakrishna M. Advanced computational approaches to understand protein aggregation. BIOPHYSICS REVIEWS 2024; 5:021302. [PMID: 38681860 PMCID: PMC11045254 DOI: 10.1063/5.0180691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 03/18/2024] [Indexed: 05/01/2024]
Abstract
Protein aggregation is a widespread phenomenon implicated in debilitating diseases like Alzheimer's, Parkinson's, and cataracts, presenting complex hurdles for the field of molecular biology. In this review, we explore the evolving realm of computational methods and bioinformatics tools that have revolutionized our comprehension of protein aggregation. Beginning with a discussion of the multifaceted challenges associated with understanding this process and emphasizing the critical need for precise predictive tools, we highlight how computational techniques have become indispensable for understanding protein aggregation. We focus on molecular simulations, notably molecular dynamics (MD) simulations, spanning from atomistic to coarse-grained levels, which have emerged as pivotal tools in unraveling the complex dynamics governing protein aggregation in diseases such as cataracts, Alzheimer's, and Parkinson's. MD simulations provide microscopic insights into protein interactions and the subtleties of aggregation pathways, with advanced techniques like replica exchange molecular dynamics, Metadynamics (MetaD), and umbrella sampling enhancing our understanding by probing intricate energy landscapes and transition states. We delve into specific applications of MD simulations, elucidating the chaperone mechanism underlying cataract formation using Markov state modeling and the intricate pathways and interactions driving the toxic aggregate formation in Alzheimer's and Parkinson's disease. Transitioning we highlight how computational techniques, including bioinformatics, sequence analysis, structural data, machine learning algorithms, and artificial intelligence have become indispensable for predicting protein aggregation propensity and locating aggregation-prone regions within protein sequences. Throughout our exploration, we underscore the symbiotic relationship between computational approaches and empirical data, which has paved the way for potential therapeutic strategies against protein aggregation-related diseases. In conclusion, this review offers a comprehensive overview of advanced computational methodologies and bioinformatics tools that have catalyzed breakthroughs in unraveling the molecular basis of protein aggregation, with significant implications for clinical interventions, standing at the intersection of computational biology and experimental research.
Collapse
Affiliation(s)
- Deepshikha Ghosh
- Department of Biological Sciences and Engineering, Indian Institute of Technology (IIT) Gandhinagar, Palaj, Gujarat 382355, India
| | - Anushka Biswas
- Department of Chemical Engineering, Indian Institute of Technology (IIT) Gandhinagar, Palaj, Gujarat 382355, India
| | | |
Collapse
|
48
|
Herlah B, Pavlin M, Perdih A. Molecular choreography: Unveiling the dynamic landscape of type IIA DNA topoisomerases before T-segment passage through all-atom simulations. Int J Biol Macromol 2024; 269:131991. [PMID: 38714283 DOI: 10.1016/j.ijbiomac.2024.131991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/09/2024] [Accepted: 04/28/2024] [Indexed: 05/09/2024]
Abstract
Type IIA DNA topoisomerases are molecular nanomachines responsible for controlling topological states of DNA molecules. Here, we explore the dynamic landscape of yeast topoisomerase IIA during key stages of its catalytic cycle, focusing in particular on the events preceding the passage of the T-segment. To this end, we generated six configurations of fully catalytic yeast topo IIA, strategically inserted a T-segment into the N-gate in relevant configurations, and performed all-atom simulations. The essential motion of topo IIA protein dimer was characterized by rotational gyrating-like movement together with sliding motion within the DNA-gate. Both appear to be inherent properties of the enzyme and an inbuilt feature that allows passage of the T-segment through the cleaved G-segment. Coupled dynamics of the N-gate and DNA-gate residues may be particularly important for controlled and smooth passage of the T-segment and consequently the prevention of DNA double-strand breaks. QTK loop residue Lys367, which interacts with ATP and ADP molecules, is involved in regulating the size and stability of the N-gate. The unveiled features of the simulated configurations provide insights into the catalytic cycle of type IIA topoisomerases and elucidate the molecular choreography governing their ability to modulate the topological states of DNA topology.
Collapse
Affiliation(s)
- Barbara Herlah
- Theory Department, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Matic Pavlin
- Department of Catalysis and Chemical Reaction Engineering, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia
| | - Andrej Perdih
- Theory Department, National Institute of Chemistry, Hajdrihova 19, 1000 Ljubljana, Slovenia; University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia.
| |
Collapse
|
49
|
Zhu Y, Zhao X, Xiang C, Liu X, Li J. Evaluation of Essential Dynamics and Fixed-Length Coarse Graining for Multidomain Proteins. J Phys Chem B 2024; 128:5147-5156. [PMID: 38758598 PMCID: PMC11619176 DOI: 10.1021/acs.jpcb.3c08198] [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: 05/19/2024]
Abstract
For multiscale modeling of biomolecules, reliable coarse-grained (CG) models can offer great potential to simulate larger temporal and spatial scales than traditional all-atom (AA) models. In this study, we explore the essential dynamics coarse graining (EDCG) and fixed-length coarse graining (FLCG) approaches for constructing highly coarse-grained models for multidomain proteins (MDPs), with 1 to 10 amino acid residues per CG site. In the studies of 13 MDPs, our data indicate that both EDCG and FLCG can preserve the protein dynamics of MDPs. FLCG, which restricts an equal number of residues in each CG site, represents an excellent approximation to EDCG and a straightforward approach for coarse-graining MDPs. Furthermore, FLCG is tested with a class B G-protein-coupled receptor protein, and the agreement with prior experiments suggests its general application to various MDPs in different environments or conditions. Finally, we demonstrate another application of FLCG through progressive backmapping, showcasing the ability to recover from lower-resolution CG models (6 residues/CG site) to higher-resolution ones (1 residue/CG site). These promising outcomes underscore the broad applicability of FLCG to construct highly or ultra-coarse-grained models of complex biomolecules for multiscale simulations.
Collapse
Affiliation(s)
- Yu Zhu
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907
| | - Xiaochuan Zhao
- Department of Chemistry, University of Vermont, Burlington, VT 05405
| | - Chijian Xiang
- Department of Horticulture & Landscape Architecture, Purdue University, West Lafayette, IN 47907
| | - Xianshi Liu
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907
| | - Jianing Li
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907
- Department of Chemistry, University of Vermont, Burlington, VT 05405
| |
Collapse
|
50
|
Ose NJ, Campitelli P, Modi T, Kazan IC, Kumar S, Ozkan SB. Some mechanistic underpinnings of molecular adaptations of SARS-COV-2 spike protein by integrating candidate adaptive polymorphisms with protein dynamics. eLife 2024; 12:RP92063. [PMID: 38713502 PMCID: PMC11076047 DOI: 10.7554/elife.92063] [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: 05/08/2024] Open
Abstract
We integrate evolutionary predictions based on the neutral theory of molecular evolution with protein dynamics to generate mechanistic insight into the molecular adaptations of the SARS-COV-2 spike (S) protein. With this approach, we first identified candidate adaptive polymorphisms (CAPs) of the SARS-CoV-2 S protein and assessed the impact of these CAPs through dynamics analysis. Not only have we found that CAPs frequently overlap with well-known functional sites, but also, using several different dynamics-based metrics, we reveal the critical allosteric interplay between SARS-CoV-2 CAPs and the S protein binding sites with the human ACE2 (hACE2) protein. CAPs interact far differently with the hACE2 binding site residues in the open conformation of the S protein compared to the closed form. In particular, the CAP sites control the dynamics of binding residues in the open state, suggesting an allosteric control of hACE2 binding. We also explored the characteristic mutations of different SARS-CoV-2 strains to find dynamic hallmarks and potential effects of future mutations. Our analyses reveal that Delta strain-specific variants have non-additive (i.e., epistatic) interactions with CAP sites, whereas the less pathogenic Omicron strains have mostly additive mutations. Finally, our dynamics-based analysis suggests that the novel mutations observed in the Omicron strain epistatically interact with the CAP sites to help escape antibody binding.
Collapse
Affiliation(s)
- Nicholas James Ose
- Department of Physics and Center for Biological Physics, Arizona State UniversityTempeUnited States
| | - Paul Campitelli
- Department of Physics and Center for Biological Physics, Arizona State UniversityTempeUnited States
| | - Tushar Modi
- Department of Physics and Center for Biological Physics, Arizona State UniversityTempeUnited States
| | - I Can Kazan
- Department of Physics and Center for Biological Physics, Arizona State UniversityTempeUnited States
| | - Sudhir Kumar
- Institute for Genomics and Evolutionary Medicine, Temple UniversityPhiladelphiaUnited States
- Department of Biology, Temple UniversityPhiladelphiaUnited States
- Center for Genomic Medicine Research, King Abdulaziz UniversityJeddahSaudi Arabia
| | - Sefika Banu Ozkan
- Department of Physics and Center for Biological Physics, Arizona State UniversityTempeUnited States
| |
Collapse
|