1
|
Clayton J, Shi L, Robertson MJ, Skiniotis G, Michaelides M, Stavitskaya L, Shen J. A putative binding model of nitazene derivatives at the μ-opioid receptor. Neuropharmacology 2025; 273:110437. [PMID: 40185362 DOI: 10.1016/j.neuropharm.2025.110437] [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: 10/03/2024] [Revised: 02/09/2025] [Accepted: 03/28/2025] [Indexed: 04/07/2025]
Abstract
Nitazenes are a class of novel synthetic opioids with exceptionally high potency. Currently, an experimental structure of μOR-opioid receptor (μOR) in complex with a nitazene is lacking. Here we used a suite of computational tools, including consensus docking, conventional molecular dynamics (MD) and metadynamics simulations, to investigate the μOR binding modes of nitro-containing meto-, eto-, proto-, buto-, and isotonitazenes and nitro-less analogs, metodes-, etodes-, and protodesnitazenes. Docking generated three binding modes, whereby the nitro-substituted or unsubstituted benzimidazole group extends into SP1 (subpocket 1 between transmembrane helix or TM 2 and 3), SP2 (subpocket 2 between TM1, TM2, and TM7) or SP3 (subpocket 3 between TM5 and TM6). Simulations suggest that etonitazene and likely also other nitazenes favor the SP2-binding mode. Comparison to the experimental structures of μOR in complex with BU72, fentanyl, and mitragynine pseudoindoxyl (MP) allows us to propose a putative model for μOR-ligand recognition in which ligand can access hydrophobic SP1 or hydrophilic SP2, mediated by the conformational change of Gln1242.60. Interestingly, in addition to water-mediated hydrogen bonds, the nitro group in nitazenes forms a π-hole interaction with the conserved Tyr751.39. Our computational analysis provides new insights into the mechanism of μOR-opioid recognition, paving the way for investigations of the structure-activity relationships of nitazenes.
Collapse
Affiliation(s)
- Joseph Clayton
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, 20993, USA; Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, NIH/DHHS, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Michael J Robertson
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Georgios Skiniotis
- Department of Molecular and Cellular Physiology, Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Michael Michaelides
- Biobehavioral Imaging & Molecular Neuropsychopharmacology Section, Neuroimaging Research Branch, National Institute on Drug Abuse, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Lidiya Stavitskaya
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug Administration, Silver Spring, MD, 20993, USA.
| | - Jana Shen
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, 21201, USA.
| |
Collapse
|
2
|
Kamel EM, Abdelrheem DA, Salah B, Lamsabhi AM. A mechanistic experimental and computational exploration of aldose reductase inhibition by coumarins from Ruta chalepensis. Biochem Biophys Res Commun 2025; 769:151946. [PMID: 40347621 DOI: 10.1016/j.bbrc.2025.151946] [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: 01/21/2025] [Revised: 05/04/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025]
Abstract
Aldose reductase (AR), a key enzyme in the polyol pathway, contributes to diabetic complications through oxidative stress and cellular damage. We investigated the inhibitory potential of coumarins from Ruta chalepensis as AR inhibitors, combining experimental and computational approaches. Among studied coumarins, aegelinol demonstrated the strongest AR inhibition with a mixed inhibition mode (IC50 = 3.75 ± 0.11 μM, Kᵢ = 3.04 ± 0.82 μM), approaching the potency of the positive control quercetin (IC50 = 2.22 ± 0.27 μM). Marmesinin and gelseminic acid also showed notable inhibition (IC50 = 5.32 ± 0.15 μM, 5.82 ± 0.61 μM, respectively), with noncompetitive binding mechanisms (Kᵢ = 4.86 ± 0.35 μM and 4.46 ± 0.52 μM), suggesting allosteric binding. Docking highlighted key interactions with critical AR residues, while molecular dynamics simulations provided insights into the stability and dynamics of AR-ligand complexes. Various MD parameters confirmed structural integrity, with the aegelinol-AR complex exhibiting the highest stability. MM/PBSA calculations supported these findings, showing the most favorable binding free energy for aegelinol, followed by marmesinin and gelseminic acid. Potential energy landscape analyses showed that aegelinol forms the most stable AR complex with the deepest energy basin. ADMET analysis confirmed aegelinol as a promising lead due to its high gastrointestinal absorption, BBB permeability, and balanced lipophilicity. Thus, Aegelinol, marmesinin, and gelseminic acid show strong potential as AR inhibitors, with aegelinol as the leading candidate for therapeutic development. These findings highlight Ruta chalepensis as a promising source of bioactive coumarins for targeted therapies against diabetic complications.
Collapse
Affiliation(s)
- Emadeldin M Kamel
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62514, Egypt.
| | - Doaa A Abdelrheem
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Bashir Salah
- Department of Industrial Engineering, College of Engineering, King Saud University, P.O. Box 800, Riyadh, 11421, Saudi Arabia
| | - Al Mokhtar Lamsabhi
- Departamento de Química, Módulo 13, Universidad Autónoma de Madrid, Campus de Excelencia UAM-CSIC Cantoblanco, 28049, Madrid, Spain; Institute for Advanced Research in Chemical Sciences (IAdChem), Universidad Autónoma de Madrid, 28049, Madrid, Spain
| |
Collapse
|
3
|
Mahto FK, Hamid I, Bhattacharya S. Molecular dynamics study of monomeric chorismate mutase shows large reduction in conformational diversity of loops upon binding of the transition state analog. J Mol Graph Model 2025; 138:109059. [PMID: 40273750 DOI: 10.1016/j.jmgm.2025.109059] [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: 02/23/2025] [Revised: 04/08/2025] [Accepted: 04/18/2025] [Indexed: 04/26/2025]
Abstract
In this in silico study, we investigated the structure and dynamics of the molten globule enzyme, monomeric chorismate mutase, which catalyzes the conversion of chorismate to prephenate despite its molten globule state. The primary aim was to understand how the enzyme stabilizes the transition state of the reaction while maintaining its molten globule characteristics. Using the transition state analog (TSA) from the NMR structure (PDB code 2GTV), molecular dynamics simulations revealed multiple hydrogen bonds between three of the enzyme's helices and the TSA. Specific residues that formed stable hydrogen bonds with the TSA were identified as potential mutation targets. Furthermore, the binding of the TSA significantly reduced the entropy of the enzyme and led to the rigidification of the backbone dihedrals across all helices. The flexibility of the loop connecting helices 1 and 2, was also analyzed, showing reduced conformational diversity upon TSA binding. Structural differences between the apo and TSA-bound forms were noted, with helices 3 and 4 exhibiting altered helicity, including a kink in helix 3 and unravelling in helix 4. Despite its molten globule nature, monomeric chorismate mutase can stabilize the TSA through hydrogen bonds involving charged residues, which are essential for maintaining the helix bundle structure. This study highlights the importance of local structural dynamics and entropy changes in enzyme catalysis, offering insights into how molten globule states can support efficient enzymatic activity.
Collapse
Affiliation(s)
- Farindra Kumar Mahto
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, 400076, India.
| | - Iqra Hamid
- Sri Pratap College, Srinagar, 190001, India.
| | - Swati Bhattacharya
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, 400076, India.
| |
Collapse
|
4
|
KT SS, Qiao B, Marmorstein JG, Wang Y, Favaro DC, Stebe KJ, Petersson EJ, Radhakrishnan R, de la Fuente‐Nunez C, Tu RS, Maldarelli C, Olvera de la Cruz M, Messinger RJ. The Role of Asparagine as a Gatekeeper Residue in the Selective Binding of Rare Earth Elements by Lanthanide-Binding Peptides. Chemistry 2025; 31:e202501318. [PMID: 40312258 PMCID: PMC12160965 DOI: 10.1002/chem.202501318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2025] [Accepted: 04/29/2025] [Indexed: 05/03/2025]
Abstract
Lanthanide-binding tag (LBT) peptides selectively complex lanthanide cations (Ln3+) in their binding pockets and are promising for lanthanide separation. However, designing LBTs that selectively target specific Ln3+ cations remains a challenge due to limited molecular-level understanding and control of interactions within the lanthanide-binding pocket. In this study, we reveal that the N5 asparagine residue acts as a gatekeeper in the binding pocket, resulting in a 100-fold selectivity for smaller Lu3+ over larger La3+ cations. Nuclear magnetic resonance spectroscopy and molecular dynamics simulations show that the N5 residue weakly binds to the larger La3+ cation, permitting H2O molecules inside the pocket. For the smaller Lu3+ cations, the N5 residue forms an inter-arm hydrogen bond with the E14 glutamic acid residue, locking the Lu3+ cation in the pocket and preventing H2O infiltration. Mutating the N5 asparagine to a D5 aspartic acid prevents such a hydrogen bond, eliminating the gatekeeping mechanism and precipitously reducing selectivity. The resulting binding affinity to Ln3+ cations is non-monotonic but generally increases with cation size. These results suggest a molecular design paradigm: the reduced affinity for larger lanthanides is due to open pocket conformations, while the selectivity of smaller Ln3+ cations over larger ones is due to the gatekeeping hydrogen bond.
Collapse
Affiliation(s)
- Surabh S. KT
- Department of Chemical EngineeringThe City College of New York, CUNY160 Convent AveNew YorkNY10031USA
| | - Baofu Qiao
- Department of Natural SciencesBaruch CollegeCity University of New York55 Lexington AveNew YorkNY10010USA
| | - Jason G. Marmorstein
- Department of ChemistryUniversity of Pennsylvania231 S 34th StPhiladelphiaPA19104USA
| | - Yiming Wang
- Department of Chemical and Biomolecular EngineeringUniversity of Pennsylvania311A, 220 S 33rd St Towne BuildingPhiladelphiaPA19104USA
- Department of BioengineeringUniversity of Pennsylvania240 Skirkanich Hall, 210 S 33rd StPhiladelphiaPA19104USA
| | - Denize C. Favaro
- Structural Biology InitiativeCUNY Advanced Science Research Center85 St Nicholas TerraceNew YorkNY10031USA
| | - Kathleen J. Stebe
- Department of Chemical and Biomolecular EngineeringUniversity of Pennsylvania311A, 220 S 33rd St Towne BuildingPhiladelphiaPA19104USA
| | - E. James Petersson
- Department of ChemistryUniversity of Pennsylvania231 S 34th StPhiladelphiaPA19104USA
| | - Ravi Radhakrishnan
- Department of Chemical and Biomolecular EngineeringUniversity of Pennsylvania311A, 220 S 33rd St Towne BuildingPhiladelphiaPA19104USA
- Department of BioengineeringUniversity of Pennsylvania240 Skirkanich Hall, 210 S 33rd StPhiladelphiaPA19104USA
| | - Cesar de la Fuente‐Nunez
- Department of Chemical and Biomolecular EngineeringUniversity of Pennsylvania311A, 220 S 33rd St Towne BuildingPhiladelphiaPA19104USA
- Department of BioengineeringUniversity of Pennsylvania240 Skirkanich Hall, 210 S 33rd StPhiladelphiaPA19104USA
- Departments of Microbiology and PsychiatryRobert Wood Johnson PavilionUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Raymond S. Tu
- Department of Chemical EngineeringThe City College of New York, CUNY160 Convent AveNew YorkNY10031USA
| | - Charles Maldarelli
- Department of Chemical EngineeringThe City College of New York, CUNY160 Convent AveNew YorkNY10031USA
| | - Monica Olvera de la Cruz
- Department of Material Science and EngineeringNorthwestern UniversityRoom 2036, Cook Hall, 2220 Campus DriveEvanstonIL60208USA
| | - Robert J. Messinger
- Department of Chemical EngineeringThe City College of New York, CUNY160 Convent AveNew YorkNY10031USA
| |
Collapse
|
5
|
Chiu S, Alexandrova AN. Cation Effects on CO 2 Delivery to Cu Electrode in Reactive Capture of CO 2. J Phys Chem Lett 2025:6032-6039. [PMID: 40489237 DOI: 10.1021/acs.jpclett.5c01022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
The direct electrochemical conversion of captured CO2, known as reactive capture of CO2 (RCC), remains a formidable challenge in heterogeneous catalysis. Given that amines are one of the most widely used capture agents for CO2, it would be desirable to electrochemically reduce the resultant adducts, such as carbamate, directly in RCC. However, current understanding suggests that the primary species undergoing reduction in RCC with amines is the CO2 dissociated from the sorbent. Herein, we employ ab initio molecular dynamics (AIMD) with DFT to analyze how the nature of alkali metal cations in the electrolyte affects carbamate at the Cu surface, thereby assessing the possibility of promoting RCC by cation effects. The simulations show that the carbamate's orientation with respect to the electrode is governed by the optimal distance between the carbamate and the cation, specifically how this distance aligns with the cation's hydration spheres. Moreover, the slow-growth AIMD results indicate that the CO2 dissociation barrier correlates with the orientation of carbamate at the interface. When the carbamate resides beyond the cation's first hydration sphere, it adopts a flat orientation with respect to the surface that promotes the release of CO2 from the capture agent. In contrast, when the carbamate disrupts the first hydration sphere and exhibits a strong cation-π interaction, it adopts an upright orientation that is less conducive to CO2 release. These findings reveal a nontrivial cation effect in RCC, suggesting that it should be possible to optimize RCC via the choice of the electrolyte.
Collapse
|
6
|
Benjamin PL, Gerhards L, Solov'yov IA, Hore PJ. Magnetosensitivity of Model Flavin-Tryptophan Radical Pairs in a Dynamic Protein Environment. J Phys Chem B 2025. [PMID: 40464336 DOI: 10.1021/acs.jpcb.5c01187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
Light-induced radical pairs in cryptochrome proteins located in the retina are thought to be the receptors at the heart of the magnetic compass sense of migratory songbirds. Reliable simulations of the performance of such sensors face several fundamental challenges. The quantum spin dynamics of large spin systems must be modeled for periods in excess of a microsecond including realistic local magnetic interactions that fluctuate on a picosecond to microsecond time scale as a result of thermal motion. Here we employ newly developed computational methods that combine explicitly time-dependent internal magnetic interactions, obtained from molecular dynamics simulations and electronic structure calculations, with efficiently and accurately modeled spin dynamics of multinuclear electron-nuclear spin systems. We identify the range of frequencies of molecular motions that are expected to have the greatest effects on the sensitivity of the proposed compass to the direction of an Earth-strength magnetic field and obtain new insights into the potential enhancements in detection sensitivity afforded by thermal modulations of electron-nuclear hyperfine interactions.
Collapse
Affiliation(s)
| | - Luca Gerhards
- Institute of Physics, Carl von Ossietzky Universität Oldenburg, Carl-von-Ossietzky Str. 9-11, 26129 Oldenburg, Germany
| | - Ilia A Solov'yov
- Institute of Physics, Carl von Ossietzky Universität Oldenburg, Carl-von-Ossietzky Str. 9-11, 26129 Oldenburg, Germany
- Research Center for Neurosensory, Science, Carl von Ossietzky Universität Oldenburg, 26111 Oldenburg, Germany
- Center for Nanoscale Dynamics (CENAD), Institute of Physics, Carl von Ossietzky Universität Oldenburg, Ammerländer Heerstr. 114-118, 26129 Oldenburg, Germany
| | - P J Hore
- Department of Chemistry, University of Oxford, Oxford OX1 3QZ, U.K
| |
Collapse
|
7
|
Millan-Pacheco C, Serratos IN, Félix-Martínez GJ, Blancas-Flores G, Osorno A, Godínez R. Cholesterol Concentration in Cell Membranes and its Impact on Receptor-Ligand Interaction: A Computational Study of ATP-Sensitive Potassium Channels and ATP Binding. J Membr Biol 2025; 258:225-236. [PMID: 40137942 DOI: 10.1007/s00232-025-00345-4] [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: 11/28/2024] [Accepted: 03/10/2025] [Indexed: 03/29/2025]
Abstract
This work describes a computer study that looks at how different amounts of cholesterol (0%, 25%, and 50%) in cell membranes change the relationship between ATP and the KATP channel. This could explain why pancreatic beta-cells secrete insulin differently. We use computer simulations of molecular dynamics, calculations of binding free energy, and an integrated oscillator model to look at the electrical activity of beta-cells. There is a need for this kind of multiscale approach right now because cholesterol plays a part in metabolic syndrome and early type 2 diabetes. Our results showed that the increase in cholesterol concentration in the cell membrane affects the electrostatic interactions between ATP and the KATP channel, especially with charged residues in the binding site. Cholesterol can influence the properties of a membrane, including its local charge distribution near the channel. This affects the electrostatic environment around the ATP-binding site, increasing the affinity of ATP for the channel as our results indicated from 0 to 25 and 50% cholesterol (- 141 to - 113 kJ/mol, respectively). Simulating this change in the affinity to ATP of the KATP channels in a model of the electrical activity of the pancreatic beta-cell indicates that even a minimal increase could produce hyperinsulism. The study answers an important research question about how the structure of the membrane affects the function of KATP and, in turn, insulin releases a common feature of metabolic syndrome and early stages of type 2 diabetes.
Collapse
Affiliation(s)
- Cesar Millan-Pacheco
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Morelos. Av. Universidad No. 1001, Colonia Chamilpa, 62209, Morelos, México
| | - Iris N Serratos
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina. Iztapalapa, C. P. 09340, Ciudad de Mexico, México.
| | - Gerardo J Félix-Martínez
- Departamento de Ingeniería Eléctrica, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina. Iztapalapa, C. P. 09340, Ciudad de Mexico, México
| | - Gerardo Blancas-Flores
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Av. San Rafael Atlixco 186, Col. Vicentina, Iztapalapa, C. P. 09340, Ciudad de Mexico, México
| | - Alejandra Osorno
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina. Iztapalapa, C. P. 09340, Ciudad de Mexico, México
| | - Rafael Godínez
- Departamento de Ingeniería Eléctrica, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina. Iztapalapa, C. P. 09340, Ciudad de Mexico, México.
| |
Collapse
|
8
|
Kamel EM, Abdelrheem DA, Salah B, Lamsabhi AM. Phytochemical inhibitors of squalene epoxidase: Integrated In silico and In vitro mechanistic insights for targeting cholesterol biosynthesis. Arch Biochem Biophys 2025; 768:110372. [PMID: 40054651 DOI: 10.1016/j.abb.2025.110372] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/17/2025] [Accepted: 03/03/2025] [Indexed: 04/15/2025]
Abstract
Squalene epoxidase (SQLE) is a critical enzyme in the sterol biosynthesis pathway and a promising therapeutic target for various diseases. This study investigates the inhibitory potential of six phytochemicals, amentoflavone, dihydromyricetin, withaferin A, ursolic acid, paeonol, and maslinic acid, against SQLE through an integrated approach combining in silico predictions and experimental validation. Computational analyses, including molecular docking, molecular dynamics (MD) simulations, Potential energy landscape (PEL), MM/PBSA analysis, and ADMET profiling, identified amentoflavone, dihydromyricetin, and withaferin A as the most promising inhibitors, with high binding affinities and stable interactions within the SQLE binding site. Among these, amentoflavone exhibited the strongest binding affinity (-10.4 kcal/mol), binding free energy (-42.01 ± 2.78 kcal/mol), and stability during a 300 ns MD simulation, supported by favorable MD trajectory and interaction energy profiles. Experimental in vitro assays further validated these findings, showing that all tested compounds exhibited inhibitory activity against SQLE, with amentoflavone demonstrating the lowest IC50 (1.92 ± 0.28 μM), confirming its role as a potent inhibitor. Enzyme kinetics studies revealed that maslinic acid and ursolic acid exhibited noncompetitive inhibition, while withaferin A, paeonol, and amentoflavone acted as competitive inhibitors. Dihydromyricetin demonstrated a mixed inhibition mode due to its dual interaction with the enzyme's active and allosteric sites. The pharmacokinetic analysis indicated that all compounds exhibited drug-like properties, with varying ADMET profiles influencing their potential as therapeutic candidates. This study highlights the therapeutic potential of amentoflavone, withaferin A, and dihydromyricetin as potent SQLE inhibitors, underscoring the value of integrated in silico and in vitro approaches in drug discovery.
Collapse
Affiliation(s)
- Emadeldin M Kamel
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62514, Egypt.
| | - Doaa A Abdelrheem
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Bashir Salah
- Department of Industrial Engineering, College of Engineering, King Saud University, P.O. Box 800, Riyadh, 11421, Saudi Arabia
| | - Al Mokhtar Lamsabhi
- Departamento de Química, Módulo 13, Universidad Autónoma de Madrid, Campus de Excelencia UAM-CSIC Cantoblanco, 28049, Madrid, Spain; Institute for Advanced Research in Chemical Sciences (IAdChem), Universidad Autónoma de Madrid, 28049, Madrid, Spain
| |
Collapse
|
9
|
Xu Y, Huang J. Quantifying the Cooperativity of Backbone Hydrogen Bonding. J Comput Chem 2025; 46:e70133. [PMID: 40387034 DOI: 10.1002/jcc.70133] [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: 04/18/2025] [Revised: 04/25/2025] [Accepted: 05/02/2025] [Indexed: 05/20/2025]
Abstract
The hydrogen bonds (H-bonds) between backbone amide and carbonyl groups are fundamental to the stability, structure, and dynamics of proteins. A key feature of such hydrogen bonding interactions is that multiple H-bonds can enhance each other when aligned, as such in theα $$ \alpha $$ -helix orβ $$ \beta $$ -sheet secondary structures. To better understand this cooperative effect, we propose a new physical quantity to evaluate the cooperativity of intermolecular interactions. Using H-bond aligned N-methylacetamide molecules as the model system, we assess the cooperativity of protein backbone hydrogen bonds using quantum chemistry (QM) calculations at the MP2/aug-cc-pVTZ level, revealing cooperative energies ranging from 2 to 4.3 kcal/mol. A set of protein force fields was benchmarked against QM results. While the additive force field failed to reproduce cooperativity, polarizable force fields, including the Drude and AMOEBA protein force fields, have been found to reproduce the trend of QM results, albeit with smaller magnitude. This work demonstrates the theoretical utility of the proposed formula for quantifying cooperativity and its relevance in force field parameterization. Incorporating cooperative energy into polarizable models presents a pathway to achieving more accurate simulations of biomolecular systems.
Collapse
Affiliation(s)
- You Xu
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Westlake AI Therapeutics Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Jing Huang
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Westlake AI Therapeutics Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| |
Collapse
|
10
|
Jadidi M, Babaali V, InanlooRahatloo K, Salehi N, Mollazadeh R. Identification of a rare variant in TNNT3 responsible for familial dilated cardiomyopathy through whole-exome sequencing and in silico analysis. Eur J Med Res 2025; 30:424. [PMID: 40437600 PMCID: PMC12117963 DOI: 10.1186/s40001-025-02692-3] [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: 10/05/2024] [Accepted: 05/15/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a prevalent etiology of heart failure, distinguished by the gradual and frequently irreversible myocardial muscle impairment. Roughly 50% of DCM occurrences stem from hereditary rare variants. In this study, our aim was to identify the genetic cause of DCM in a pedigree with several affected individuals across four generations. METHODS Whole exome sequencing was performed on the proband, with variants filtered and analyzed using in silico tools. Co-segregation analysis was conducted using Sanger sequencing. Protein structure modeling and protein-protein interaction evaluations were performed using AlphaFold3 and HADDOCK2.4, respectively. RESULTS We identified a missense rare variant in the TNNT3 gene, leading to the p.Glu125Gly alteration in the Troponin T3 (TNNT3). This rare variant is strongly implicated as the causative factor for DCM in the pedigree. Several key factors underscore its significance: the rare variant co-segregates with the disease in the pedigree, is absent in 850 control samples, alters a conserved amino acid, is predicted to detrimentally affect protein function, and results in structural changes. CONCLUSIONS Our findings suggest that TNNT3 rare variants can induce DCM by weakening the binding energy between TNNT3 and Tropomyosin (TPM), leading to functional deficiencies in muscle contraction, as demonstrated by our structural modeling and docking studies. Troponin T is essential for the proper contraction of striated muscles and is related to cardiac development. Bioinformatics investigations have elucidated the involvement of TNNT3-related pathways, notably the Striated Muscle Contraction pathway and Cardiac Conduction. TNNT3 resides within loci previously implicated in cardiomyopathy. Given its crucial role in muscle contractile function, rare variants in TNNT3 hold the potential to be a significant contributing factor in the pathogenesis of DCM. A wealth of literature substantiates the correlation between troponin T and cardiac disorders. Our findings further corroborate this association.
Collapse
Affiliation(s)
- Motahareh Jadidi
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Vida Babaali
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Kolsoum InanlooRahatloo
- Department of Cell and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Najmeh Salehi
- Department of Applied Biological Sciences, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Reza Mollazadeh
- Department of Cardiology, School of Medicine, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Sun Q, Wang H, Xie J, Wang L, Mu J, Li J, Ren Y, Lai L. Computer-Aided Drug Discovery for Undruggable Targets. Chem Rev 2025. [PMID: 40423592 DOI: 10.1021/acs.chemrev.4c00969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Undruggable targets are those of therapeutical significance but challenging for conventional drug design approaches. Such targets often exhibit unique features, including highly dynamic structures, a lack of well-defined ligand-binding pockets, the presence of highly conserved active sites, and functional modulation by protein-protein interactions. Recent advances in computational simulations and artificial intelligence have revolutionized the drug design landscape, giving rise to innovative strategies for overcoming these obstacles. In this review, we highlight the latest progress in computational approaches for drug design against undruggable targets, present several successful case studies, and discuss remaining challenges and future directions. Special emphasis is placed on four primary target categories: intrinsically disordered proteins, protein allosteric regulation, protein-protein interactions, and protein degradation, along with discussion of emerging target types. We also examine how AI-driven methodologies have transformed the field, from applications in protein-ligand complex structure prediction and virtual screening to de novo ligand generation for undruggable targets. Integration of computational methods with experimental techniques is expected to bring further breakthroughs to overcome the hurdles of undruggable targets. As the field continues to evolve, these advancements hold great promise to expand the druggable space, offering new therapeutic opportunities for previously untreatable diseases.
Collapse
Affiliation(s)
- Qi Sun
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan 610213, China
| | - Hanping Wang
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Juan Xie
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Liying Wang
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Junxi Mu
- Peking-Tsinghua Center for Life Science, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Junren Li
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yuhao Ren
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Luhua Lai
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Science, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan 610213, China
- Research Unit of Drug Design Method, Chinese Academy of Medical Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
12
|
Zhang R, Wang Y, Wang X, Luo H, Wang Y, Yao B, Huang H, Tian J, Guan F. Influence of mutations at different distances from the active center on the activity and stability of laccase 13B22. BIORESOUR BIOPROCESS 2025; 12:47. [PMID: 40423903 PMCID: PMC12116972 DOI: 10.1186/s40643-025-00893-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/26/2025] [Accepted: 05/17/2025] [Indexed: 05/28/2025] Open
Abstract
Laccases with high catalytic efficiency and high thermostability can drive a broader application scope. However, the structural distribution of key amino acids capable of significantly influencing the performance of laccases has not been explored in depth. Thirty laccase 13B22 mutants with changes in amino acids at distances of 5 Å (first shell), 5-8 Å (second shell), and 8-12 Å (third shell) from the active center were validated experimentally with 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) as substrate. Twelve of these mutants (first shell, 1; second shell, 4; third shell, 7) showed higher catalytic efficiency than the wild-type enzyme. Mutants D511E and I88L-D511E showed 5.36- and 10.58-fold increases in kcat/Km, respectively, with increases in optimal temperature of 15 °C and optimal pH from 7.0 to 8.0. Furthermore, both mutants exhibited greater thermostability compared to the wild-type, with increases of 3.33 °C and 5.06 °C in Tm and decreases of 0.39 J and 0.59 J in total structure energy, respectively. The D511E mutation resides in the third shell, while I88L is in the second shell, and their performance enhancements were attributed to alterations in the rigidity or flexibility of specific protein structural domains. Both mutants showed enhanced degradation efficiency for benzo[a]pyrene and zearalenone. These findings highlight the importance of the residues located far from the active center in the function of laccase (second shell and third shell), suggesting broader implications for enzyme optimization and biotechnological applications.
Collapse
Affiliation(s)
- Ruohan Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yuchen Wang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Xiaolu Wang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Huiying Luo
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Yuan Wang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Bin Yao
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Huoqing Huang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - Jian Tian
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| | - Feifei Guan
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| |
Collapse
|
13
|
Hui C, de Vries R, Kopec W, de Groot BL. Effective polarization in potassium channel simulations: Ion conductance, occupancy, voltage response, and selectivity. Proc Natl Acad Sci U S A 2025; 122:e2423866122. [PMID: 40392847 DOI: 10.1073/pnas.2423866122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 04/07/2025] [Indexed: 05/22/2025] Open
Abstract
Potassium (K+) channels are widely distributed in many types of organisms. They combine high efficiency (~100 pS) and K+/Na+ selectivity by a conserved selectivity filter (SF). Molecular Dynamics (MD) simulations can provide detailed, atomistic mechanisms of this sophisticated ion permeation. However, currently there are clear inconsistencies between computational predictions and experimental results. First, the ion occupancy of the SF in simulations is lower than expected (~2.5 in MD compared to ~4 in X-ray crystallography). Second, in many reported MD simulations of K+ channels, K+ conductance is typically an order of magnitude lower than experimental values. This discrepancy is in part because the force fields used in MD simulations of potassium channels do not account for polarization. One of the proposed solutions is the Electronic Continuum Correction (ECC), a force field modification that scales down formal charges, to introduce the polarization in a mean-field way. When the ECC is used in conjunction with the Charmm36m force field, the simulated K+ conductance increases 13-fold. Following the analysis of ion occupancy states using Hamiltonian Replica Exchange simulations, we propose a parameter set for Amber14sb, that also leads to a similar increase in conductance. These two force fields are then used to compute the full current-voltage (I-V) curves from MD simulations, approaching quantitative agreement with experiments at all voltages. In general, the ECC-enabled simulations are in excellent agreement with experiment, in terms of ion occupancy, conductance, current-voltage response, and K+/Na+ selectivity.
Collapse
Affiliation(s)
- Chenggong Hui
- Computational Biomolecular Dynamics Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Reinier de Vries
- Computational Biomolecular Dynamics Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| | - Wojciech Kopec
- Computational Biomolecular Dynamics Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
- Department of Chemistry, Queen Mary University of London, London E1 4NS, United Kingdom
| | - Bert L de Groot
- Computational Biomolecular Dynamics Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37077, Germany
| |
Collapse
|
14
|
Fu L, Fu B, Zhang DH. Fundamental invariant-neural network as a correction to the intramolecular force field illustrated for the full-dimensional potential energy surface of propane. Phys Chem Chem Phys 2025. [PMID: 40424012 DOI: 10.1039/d5cp00599j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
As a highly effective approach for constructing potential energy surfaces (PESs) with both precision and efficiency, Δ-machine learning has been widely used in PES development. Inspired by the Δ-machine learning framework, we develop a combined model of fundamental invariant-neural network (FI-NN) and force field. Fitting the difference between the force field and ab initio energy by the FI-NN method is able to improve the accuracy of the force field. We demonstrate this enhanced methodology through the development of an intramolecular force field for propane, where CCSD(T)-F12a/AVTZ energies are initially approximated by the force field and subsequently refined using the FI-NN approach. Compared to the PES fitted by FI-NN, this combined method reduces the root mean square error (RMSE) by 50%.
Collapse
Affiliation(s)
- Liangfei Fu
- State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China.
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Bina Fu
- State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China.
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Dong H Zhang
- State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, P. R. China.
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
15
|
Kamel EM, Allam AA, Rudayni HA, Alkhedhairi S, Alkhayl FFA, Alwaili MA, Lamsabhi AM. Mechanistic Insights into Polyphenols-mediated Squalene Epoxidase Inhibition: Computational Models and Experimental Validation for Targeting Cholesterol Biosynthesis. Cell Biochem Biophys 2025:10.1007/s12013-025-01784-5. [PMID: 40418424 DOI: 10.1007/s12013-025-01784-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2025] [Indexed: 05/27/2025]
Abstract
Squalene epoxidase is a key enzyme in sterol biosynthesis, particularly in cholesterol metabolism. Its inhibition has emerged as a promising therapeutic strategy for metabolic disorders, hypercholesterolemia, and certain infections. Herein, we investigated the SQLE inhibitory potential of six polyphenolic compounds, identified through in silico virtual screening of a large natural phenolic library and selected for high predicted binding affinity and structural diversity. Molecular docking demonstrated strong interactions between these candidates and SQLE, with curcumin exhibiting the highest binding affinity (-10.1 kcal/mol). Molecular dynamics simulations confirmed stable interactions for all compounds, highlighting curcumin, piceatannol, and pterostilbene as particularly favorable. Their strong binding free energies were further supported by MM/PBSA calculations (-36.62 ± 4.17, -31.32 ± 3.77, and -32.01 ± 1.34 kcal/mol, respectively), corroborated by free energy landscape analysis. ADMET profiling revealed diverse pharmacokinetic properties among the six polyphenolics. In vitro testing confirmed curcumin as the most potent inhibitor (IC50 = 1.88 ± 0.21 µM), with piceatannol (2.55 ± 0.30 µM) and pterostilbene (2.69 ± 0.11 µM) following closely. Enzyme kinetics demonstrated that these three compounds act as competitive inhibitors targeting the enzyme's active site. Collectively, these findings highlight the combined power of computational and experimental approaches for identifying novel SQLE inhibitors.
Collapse
Affiliation(s)
- Emadeldin M Kamel
- Chemistry Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt.
| | - Ahmed A Allam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Hassan A Rudayni
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Saleh Alkhedhairi
- Department of Medical Biosciences, College of Veterinary Medicine, Qassim University, P.O. Box 6622, Buraidah, Saudi Arabia
| | - Faris F Aba Alkhayl
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Maha A Alwaili
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. BOX 84428, Riyadh, Saudi Arabia
| | - Al Mokhtar Lamsabhi
- Departamento de Química and Institute for advanced research in chemical Science (IAdChem), Facultad de Ciencias, Módulo 13, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
16
|
Chen C, Zhang L. Effects of structural parameters on the desalination performance of a multilayer stacked graphene oxide membrane: insights from molecular dynamics simulation. NANOSCALE 2025; 17:12868-12879. [PMID: 40326607 DOI: 10.1039/d5nr00314h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
With the rapid development of nanotechnology, functionalized graphene membranes have shown great advantages, attracting increasing attention in recent years. However, the effects of different groups on the desalination performance of layered graphene membranes, as well as the transport and separation mechanisms of water molecules and ions within two-dimensional nanochannels, remain insufficiently understood. In this study, molecular dynamics simulations are employed to investigate the influence of multiple factors on seawater desalination performance, focusing on transport behavior and intermolecular interactions, to elucidate the structure-performance relationship between the membrane architecture and separation efficiency. Our findings indicate that the desalination performance of a multilayer stacked graphene oxide membrane (MGOM) is affected by key structural parameters, including interlayer spacing (H), offset (O), and slit width (dG). Optimal performance is achieved when H = 0.8 nm, dG = 1.1 nm, and O = 1.26 nm. Interestingly, the rejection rate of Na+ is slightly lower than that of Cl-, mainly due to the differences in the interaction between different ions and functional groups in the sheets and the hydration radius.
Collapse
Affiliation(s)
- Chen Chen
- School of Chemistry and Chemical Engineering, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou, 310018, People's Republic of China.
| | - Li Zhang
- School of Chemistry and Chemical Engineering, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou, 310018, People's Republic of China.
| |
Collapse
|
17
|
Huang CH, Lewis R, Thomas S, Tang Z, Jones J, Nason S, Zuverza-Mena N, Piskulich ZA, O'Keefe TL, Tuga B, Paredes-Beaulieu A, Vasiliou V, Cui Q, Dalluge JJ, White JC, Haynes CL. Designing Ultraporous Mesostructured Silica Nanoparticles for the Remediation of Per- and Polyfluoroalkyl Substances. ACS NANO 2025. [PMID: 40402145 DOI: 10.1021/acsnano.5c02008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2025]
Abstract
Concerns about per- and polyfluoroalkyl substances (PFAS) have been raised globally as they are bioaccumulative, highly persistent, and invoke a range of health risks. Although phytoremediation is a sustainable PFAS remediation strategy, its efficiency is highly dependent on the PFAS analyte chain length, with limited uptake and removal of longer-chain contaminants. This study aims to develop surface-modified ultraporous mesostructured silica nanoparticles (UMNs) to facilitate PFAS phytoremediation. UMNs were synthesized and functionalized to tune their hydrophobicity and surface charge to enhance UMN affinity for PFAS. Dynamic light scattering, ς-potential, and nitrogen physisorption show that the modified UMNs had similar physical characteristics. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis shows that positively charged UMNs have a higher affinity for PFAS than negatively charged UMNs (with 20% of perfluorooctanoic acid, or PFOA, remaining in solution vs 100% of PFOA remaining in solution, respectively). When incubated with multiple PFAS, UMNs show greater removal efficiency for longer-chain and more hydrophobic PFAS. Preliminary plant studies in soil show an increased PFOA bioconcentration when positively charged UMNs are present. Molecular dynamics simulations, which focused on interactions between the different functional groups on the silica surface and PFAS molecules, were completed and show the importance of the combination of hydrophobic and electrostatic interactions to drive PFAS uptake. Overall, this study highlights the potential of surface-modified UMNs to enhance the uptake of PFAS from the environmental matrix and promote phytoremediation.
Collapse
Affiliation(s)
- Cheng-Hsin Huang
- Department of Chemistry, University of Minnesota-Twin Cities, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Riley Lewis
- Department of Chemistry, University of Minnesota-Twin Cities, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Sara Thomas
- The Connecticut Agricultural Experiment Station, 123 Huntington Street, New Haven, Connecticut 06511, United States
| | - Zhengyi Tang
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Jasmine Jones
- The Connecticut Agricultural Experiment Station, 123 Huntington Street, New Haven, Connecticut 06511, United States
| | - Sara Nason
- The Connecticut Agricultural Experiment Station, 123 Huntington Street, New Haven, Connecticut 06511, United States
| | - Nubia Zuverza-Mena
- The Connecticut Agricultural Experiment Station, 123 Huntington Street, New Haven, Connecticut 06511, United States
| | - Zeke A Piskulich
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
- Department of Quantitative Biomedicine, Rutgers University, Piscataway, New Jersey 08854, United States
| | - Tana L O'Keefe
- Department of Chemistry, University of Minnesota-Twin Cities, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Beza Tuga
- Department of Chemistry, University of Minnesota-Twin Cities, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Antavia Paredes-Beaulieu
- Department of Chemistry, University of Minnesota-Twin Cities, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut 06520, United States
| | - Qiang Cui
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Joseph J Dalluge
- Department of Chemistry, University of Minnesota-Twin Cities, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Jason C White
- The Connecticut Agricultural Experiment Station, 123 Huntington Street, New Haven, Connecticut 06511, United States
| | - Christy L Haynes
- Department of Chemistry, University of Minnesota-Twin Cities, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
18
|
Mirakhory M, Majumdar A, Ihme M, van Duin ACT. Iodine recombination in xenon solvent: Clusters in the gas to liquid-like state transition. J Chem Phys 2025; 162:194309. [PMID: 40387769 DOI: 10.1063/5.0260087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/28/2025] [Indexed: 05/20/2025] Open
Abstract
Supercritical fluids (SCFs) have attracted significant attention as solvents for chemical reactions due to their unique properties, such as high diffusivity, low viscosity, and tunable solvation properties. These properties profoundly influence reaction kinetics and are often attributed to the formation of molecular clusters within SCFs. To study the effect of supercritical solvent on chemical reactivity and dynamics of reactions, one needs to understand the dynamics of clusters in supercritical fluid. Extensive experiments on the photodissociation and recombination of iodine in supercritical fluids served as a model system for understanding these effects. Experimental studies have been complemented by theoretical and computational investigations, which mostly employ Monte Carlo or empirical molecular dynamics simulations. However, computational studies using non-reactive force fields and ab initio approaches present challenges in capturing reactive processes at larger scales within supercritical fluids. In this work, we developed the ReaxFF parameters by training against quantum mechanics data. ReaxFF reactive force field based molecular dynamics simulations were performed, studying the dynamics of a xenon solvent and cage effect at different thermodynamic conditions for the iodine recombination reaction. We show that the conditions near the critical point are the optimal conditions to study the cage effect. We show that the average lifetime of xenon clusters ranging between 5 and 11 ps is comparable to iodine geminate recombination. Our simulation results of iodine recombination in xenon solvent demonstrate the higher probability of iodine molecule formation in the presence of xenon clusters. Finally, we show that the supercritical condition exhibits the highest recombination rate for iodine atoms.
Collapse
Affiliation(s)
- M Mirakhory
- Department of Mechanical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802, USA
| | - A Majumdar
- Department of Mechanical Engineering, Stanford University, Stanford, California 94305, USA
| | - M Ihme
- Department of Mechanical Engineering, Stanford University, Stanford, California 94305, USA
| | - A C T van Duin
- Department of Mechanical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802, USA
| |
Collapse
|
19
|
Durer ZA, İnce Hİ, Düvenci ZŞ, Timuçin E, Gräwert T, Orun O, Kan B, Sayers Z. Insights into the solution structure of the actin-binding tail domain of metavinculin by small angle X-ray scattering and molecular dynamics simulations. Int J Biol Macromol 2025:144376. [PMID: 40409637 DOI: 10.1016/j.ijbiomac.2025.144376] [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: 02/27/2025] [Revised: 05/07/2025] [Accepted: 05/17/2025] [Indexed: 05/25/2025]
Abstract
Vinculin is a ubiquitously expressed focal adhesion protein that plays an important role in cell-matrix and cell-to-cell junctions. Metavinculin, a muscle-specific splice variant of vinculin, contains a 68-amino acid disordered insert region in its actin binding tail domain (MVt). Mutations in this insert are linked to cardiomyopathies. This study investigates the solution structures and structural ensembles of wild-type (WT) and two mutant MVts, ΔLeu954 and R975W, which have been associated with cardiomyopathies, using small-angle X-ray scattering (SAXS) and molecular dynamics (MD) simulations. SAXS analyses revealed subtle differences in the estimated maximum dimensions and corroborated the elongated shape of the MVts. Quantitative comparisons of SAXS profiles indicated similarity between the WT and ΔLeu954, whereas R975W exhibited differences in the small-angle region. MD simulations demonstrated reduced conformational flexibility and greater packing of the insert in WT compared to mutants. Notably, a salt-bridge observed between R975 and D928 in a WT simulation provides a structural basis for the destabilization caused by the R975W mutation. These findings provide insights into the structure and dynamics of WT and mutant MVt, reflecting the promise of SAXS combined with MD simulations to elucidate the structural properties of proteins with structural disorder.
Collapse
Affiliation(s)
- Zeynep Aslıhan Durer
- School of Medicine, Department of Biophysics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey; Faculty of Pharmacy, Department of Biochemistry, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.
| | - Hande İpek İnce
- Institute of Health Sciences, Department of Biophysics, Marmara University, Istanbul, Turkey
| | - Zeynep Şevval Düvenci
- Institute of Health Sciences, Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Emel Timuçin
- Institute of Health Sciences, Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey; School of Medicine, Department of Biostatistics and Medical Informatics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Tobias Gräwert
- European Molecular Biology Laboratory - Hamburg Unit, Biological Small Angle Scattering Group, Hamburg, Germany
| | - Oya Orun
- Institute of Health Sciences, Department of Biophysics, Marmara University, Istanbul, Turkey; School of Medicine, Department of Biophysics, Marmara University, Istanbul, Turkey
| | - Beki Kan
- School of Medicine, Department of Biophysics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Zehra Sayers
- Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics and Bioengineering Program, Sabanci University, İstanbul, Turkey
| |
Collapse
|
20
|
Shin S, Mugnai ML, Thirumalai D. Water-Mediated Interactions between Glycans Are Weakly Repulsive and Unexpectedly Long-Ranged. J Am Chem Soc 2025; 147:17448-17458. [PMID: 40357734 DOI: 10.1021/jacs.5c04126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Glycans on the cell surface play an essential role in mediating cell-cell interactions and immune response. Despite their importance, the interactions between them have not been fully characterized. Here, we reveal, using all-atom molecular dynamics simulations and free energy calculations, that water-mediated interactions between a pair of N-glycans without a net charge are weakly repulsive with a range that exceeds their sizes. Unexpectedly, the effective glycan-glycan interactions decay logarithmically as the separation between them increases. Strikingly, this finding coincides exactly with the predicted interaction, which is entropic in origin, between two star polymers consisting of long flexible polymers grafted onto colloidal particles. The weak repulsive interaction, which extends beyond the size of a glycan, is sensitive to the relative orientation of the glycans. The effective long-range repulsive interaction vanishes if the charges on water are turned off, thus establishing that electrostatic interactions, arising in part due to the persistent hydrogen bonds between water and the glycans, are responsible for the interglycan repulsion.
Collapse
Affiliation(s)
- Sucheol Shin
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Mauro L Mugnai
- Institute for Soft Matter Synthesis and Metrology, Georgetown University, Washington, District of Columbia 20057, United States
| | - D Thirumalai
- Department of Chemistry, The University of Texas at Austin, Austin, Texas 78712, United States
- Department of Physics, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
21
|
Li D, Chen H, Vale G, Elghobashi-Meinhardt N, Hatton A, Rong S, McDonald JG, Li X. Molecular insights into human phosphatidylserine synthase 2 and its regulation of SREBP pathways. Proc Natl Acad Sci U S A 2025; 122:e2501177122. [PMID: 40372437 PMCID: PMC12107096 DOI: 10.1073/pnas.2501177122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 04/17/2025] [Indexed: 05/16/2025] Open
Abstract
Homologous proteins share similar sequences, enabling them to work together in cells to support normal physiological functions. Phosphatidylserine synthases 1 and 2 (PSS1 and PSS2) are homologous enzymes that catalyze the synthesis of phosphatidylserine (PS) from different substrates. PSS2 shows a preference for phosphatidylethanolamine (PE) as its substrate, whereas PSS1 can utilize either PE or phosphatidylcholine. Previous studies showed that inhibiting PSS1 promotes SREBP-2 cleavage. Interestingly, despite their homology, our findings reveal that PSS2 exerts an opposing effect on the cleavage of both SREBP-1 and SREBP-2. We resolved the cryo-electron microscopy (cryo-EM) structure of human PSS2 at 3.3 Å resolution. Structural comparison of the catalytic cavities between PSS1 and PSS2 along with molecular dynamics simulations uncovers the molecular details behind the substrate preference of PSS2 for PE. The lipidomic analysis showed that PSS2 deficiency leads to PE accumulation in the endoplasmic reticulum, which has been shown to inhibit the cleavage of sterol regulatory element-binding proteins (SREBPs) in mice. Thus, our findings reveal the intricate network of intracellular phospholipid metabolism and underscore the distinct regulatory roles of homologous proteins in cellular activities.
Collapse
Affiliation(s)
- Dongyu Li
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Hongwen Chen
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Goncalo Vale
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX75390
| | | | - Alexandra Hatton
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Shunxing Rong
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Jeffrey G. McDonald
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX75390
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Xiaochun Li
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX75390
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX75390
| |
Collapse
|
22
|
Schwartz R, Hadar-Volk A, Nam K, Major DT. Template-Based Docking Using Automated Maximum Common Substructure Identification with EnzyDock: Mechanistic and Inhibitor Docking. J Chem Inf Model 2025. [PMID: 40388499 DOI: 10.1021/acs.jcim.5c00201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2025]
Abstract
EnzyDock is a multistate, multiscale CHARMM-based docking program which enables mechanistic docking, i.e., modeling enzyme reactions by docking multiple reaction states, like substrates, intermediates, transition states, and products to the enzyme, in addition to standard protein-ligand docking. To achieve docking of multiple reaction states with similar poses (i.e., consensus docking), EnzyDock employs consensus pose restraints of the docked ligand states relative to a docking template. In the current work, we present an implementation of a Maximum Common Substructure (MCS)-guided docking strategy using EnzyDock, enabling the automatic detection of similarity among query ligands. Specifically, the MCS multistate approach is employed to efficiently dock ligands along enzyme reaction coordinates, including reactants, intermediates, and products, which allows efficient and robust mechanistic docking. To demonstrate the effectiveness of the MCS strategy in modeling enzymes, it is first applied to two highly complex enzyme reaction cascades catalyzed by the diterpene synthase CotB2 and the Diels-Alderase LepI. In addition, the MCS strategy is applied to dock enzyme inhibitors using cocrystallized inhibitors or substrates to guide the docking in the enzymes dihydrofolate reductase and the SARS-CoV-2 enzyme Mpro. The latter case exemplifies the use of MCS with EnzyDock's covalent docking capabilities and QM/MM scoring option. We show that different protocols of the implemented MCS algorithm are needed to obtain mechanistic consistency (i.e., similar poses) in mechanistic docking or to accurately dock chemically diverse ligands in inhibitor docking. Although the current implementation is specific for EnzyDock, the findings should be general and transferable to additional docking programs.
Collapse
Affiliation(s)
- Renana Schwartz
- Department of Chemistry, Israel National Institute of Energy Storage (INIES) and Institute for Nanotechnology & Advanced Materials, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Amit Hadar-Volk
- Department of Chemistry, Israel National Institute of Energy Storage (INIES) and Institute for Nanotechnology & Advanced Materials, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Kwangho Nam
- Department of Chemistry and Biochemistry and Division of Data Science, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Dan T Major
- Department of Chemistry, Israel National Institute of Energy Storage (INIES) and Institute for Nanotechnology & Advanced Materials, Bar-Ilan University, Ramat-Gan 5290002, Israel
| |
Collapse
|
23
|
Procacci P. PDBrestore: A Free Web Interface for Processing and Fixing Protein Chains From Raw PDB Files. J Comput Chem 2025; 46:e70124. [PMID: 40365838 PMCID: PMC12076534 DOI: 10.1002/jcc.70124] [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: 02/26/2025] [Revised: 04/06/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025]
Abstract
We present PDBrestore, a free web interface for repairing protein PDB chains extracted from either a local PDB file or a PDB file downloaded from the Protein Data Bank. PDBrestore performs several key tasks: It adds hydrogen atoms, completes missing atoms in side chains, fills gaps in the sequence, derives the itp parameter file for a ligand according to the GAFF2 force field for GROMACS applications, and generates a reasonably pre-equilibrated solvated simulation box. The interface is designed to streamline the cumbersome preparatory work required to set up an initial protein-ligand coordinates PDB file for use in drug design projects, such as free energy perturbation or thermodynamic integration calculations of ligand binding affinities. Additionally, PDBrestore is available as a command-line application within the open-source ORAC distribution, which can be freely downloaded from the website: www1.chim.unifi.it/orac.
Collapse
Affiliation(s)
- Piero Procacci
- Department of ChemistryUniversity of FlorenceSesto FiorentinoItaly
| |
Collapse
|
24
|
Hozana GN, Díaz Mirón G, Hassanali A. Data-Driven Discovery of the Origins of UV Absorption in the Alpha-3C Protein. J Phys Chem B 2025; 129:4728-4737. [PMID: 40312142 DOI: 10.1021/acs.jpcb.5c00532] [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: 05/03/2025]
Abstract
Over the past decade, there has been a growing body of experimental work showing that proteins devoid of aromatic and conjugated groups can absorb light in the near-UV beyond 300 nm and emit visible light. Understanding the origins of this phenomenon offers the possibility of designing noninvasive spectroscopic probes for local interactions in biological systems. It was recently found that the synthetic protein α3C displays UV-vis absorption between 250 and 800 nm, which was shown to arise from charge-transfer excitations between charged amino acids. In this work, we use data-driven approach to re-examine the origins of these features using a combination of molecular dynamics and excited-state simulations. Specifically, an unsupervised learning approach beginning with encoding protein environments with local atomic descriptors is employed to automatically detect relevant structural motifs. We identify three main motifs corresponding to different hydrogen-bonding patterns that are subsequently used to perform QM/MM simulations, including the entire protein and solvent bath with the density-functional tight-binding (DFTB) approach. Hydrogen-bonding structures involving arginine and carboxylate groups appear to be the most prone to near-UV absorption. We show that the magnitude of the UV-vis absorption predicted from the simulations is rather sensitive to the size of the QM region employed as well as to the inclusion of explicit solvation.
Collapse
Affiliation(s)
- Germaine Neza Hozana
- International Centre for Theoretical Physics (ICTP), Strada Costiera 11, Trieste 34151, Italy
- Dipartimento di Fisica, Universitá degli Studi di Trieste, Via Alfonso Valerio 2, Trieste 34127, Italy
| | - Gonzalo Díaz Mirón
- International Centre for Theoretical Physics (ICTP), Strada Costiera 11, Trieste 34151, Italy
| | - Ali Hassanali
- International Centre for Theoretical Physics (ICTP), Strada Costiera 11, Trieste 34151, Italy
| |
Collapse
|
25
|
Santos D, Foglia L, Kiser PD, Yu A. The molecular mechanisms of visual chromophore release from cellular retinaldehyde-binding protein. Structure 2025:S0969-2126(25)00174-1. [PMID: 40403716 DOI: 10.1016/j.str.2025.04.018] [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: 11/11/2024] [Revised: 02/11/2025] [Accepted: 04/25/2025] [Indexed: 05/24/2025]
Abstract
Cellular retinaldehyde-binding protein (CRALBP) is an 11-cis-retinoid binding protein operating within the visual cycle. CRALBP serves as the terminal acceptor of 11-cis-retinaldehyde (11cRAL) produced within the retinal pigment epithelium (RPE) and mediates 11cRAL transport to the RPE apical microvilli. Crystallographic structures of CRALBP revealed that the 11cRAL-binding pocket is sealed off from bulk solvent, indicating a necessity for conformational changes to allow ligand egress. Here, we performed long timescale all-atom molecular dynamics simulations of CRALBP to elucidate the mechanisms of ligand release. CRALBP exhibits slower diffusive behavior in the presence of membranes containing negatively charged phospholipids, which bind to an exposed cationic pocket in CRALBP. Umbrella sampling calculations revealed thermodynamically likely pathways for 11cRAL egress. Our data suggest that the CRALBP-acidic phospholipid interaction facilitates 11cRAL release through allosteric, conformational changes that perturb the binding site, lowering ligand affinity. These findings offer insights into the molecular pathology of CRALBP-associated retinopathy.
Collapse
Affiliation(s)
- Daniel Santos
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92617, USA
| | - Lorenzo Foglia
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92617, USA
| | - Philip D Kiser
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92617, USA; Department of Ophthalmology, University of California, Irvine, Irvine, CA 92617, USA; Department of Clinical Pharmacy Practice, University of California, Irvine, Irvine, CA 92697, USA; Research Service, VA Long Beach Healthcare System, Long Beach, CA 92697, USA.
| | - Alvin Yu
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92617, USA.
| |
Collapse
|
26
|
Nadeem H, Shukla D. Ensemble Adaptive Sampling Scheme: Identifying an Optimal Sampling Strategy via Policy Ranking. J Chem Theory Comput 2025; 21:4626-4639. [PMID: 40261689 DOI: 10.1021/acs.jctc.4c01488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Efficient sampling in biomolecular simulations is critical for accurately capturing the complex dynamic behaviors of biological systems. Adaptive sampling techniques aim to improve efficiency by focusing computational resources on the most relevant regions of the phase space. In this work, we present a framework for identifying the optimal sampling policy through metric-driven ranking. Our approach systematically evaluates the policy ensemble and ranks the policies based on their ability to explore the conformational space effectively. Through a series of biomolecular simulation case studies, we demonstrate that the choice of a different adaptive sampling policy at each round significantly outperforms single policy sampling, leading to faster convergence and improved sampling performance. This approach takes an ensemble of adaptive sampling policies and identifies the optimal policy for the next round based on current data. Beyond presenting this ensemble view of adaptive sampling, we also propose two sampling algorithms that approximate this ranking framework on the fly. The modularity of this framework allows incorporation of any adaptive sampling policy, making it versatile and suitable as a comprehensive adaptive sampling scheme.
Collapse
Affiliation(s)
- Hassan Nadeem
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Diwakar Shukla
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Center for Biophysics and Quantitative Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
27
|
Qin Y, Mu L, Wan X, Zong Z, Li T, Fang H, Yang N. Deep Potential for Interaction between Hydrated Cs + and Graphene. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:11506-11514. [PMID: 40300096 DOI: 10.1021/acs.langmuir.5c00508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
The influence of hydrated cation-π interaction forces on the adsorption and filtration capabilities of graphene-based membrane materials is significant. However, the lack of interaction potential between hydrated Cs+ and graphene limits the scope of adsorption studies. Here, it is developed that a deep neural network potential function model to predict the interaction force between hydrated Cs+ and graphene. The deep potential has DFT-level accuracy, enabling accurate property prediction. This deep potential is employed to investigate the properties of the graphene surface solution, including the vibrational power spectrum of water density distribution, radial distribution function, and mean square displacement. Furthermore, the adsorption energy and charge between hydrated Cs+ and graphene were calculated for varying amounts of bound water, indicating that the presence of water molecules weakens the interaction between the ions and graphene. The method provides a powerful tool to study the adsorption behavior of hydrated cations on graphene surfaces and offers a new solution for handling radionuclides.
Collapse
Affiliation(s)
- Yangjun Qin
- School of Energy and Power Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Department of Physics, National University of Defense Technology, Changsha 410073, China
| | - Liuhua Mu
- School of Physical Science and Technology, Ningbo University, Ningbo 315211, China
| | - Xiao Wan
- Key Laboratory for Thermal Science and Power Engineering of Ministry of Education, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Zhicheng Zong
- School of Energy and Power Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Department of Physics, National University of Defense Technology, Changsha 410073, China
| | - Tianhao Li
- School of Energy and Power Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
- Department of Physics, National University of Defense Technology, Changsha 410073, China
| | - Haisheng Fang
- School of Energy and Power Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Nuo Yang
- Department of Physics, National University of Defense Technology, Changsha 410073, China
| |
Collapse
|
28
|
Cetin E, Abdizadeh H, Atilgan AR, Atilgan C. A Thermodynamic Cycle to Predict the Competitive Inhibition Outcomes of an Evolving Enzyme. J Chem Theory Comput 2025; 21:4910-4920. [PMID: 40268874 PMCID: PMC12080111 DOI: 10.1021/acs.jctc.5c00193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Understanding competitive inhibition at the molecular level is essential for unraveling the dynamics of enzyme-inhibitor interactions and predicting the evolutionary outcomes of resistance mutations. In this study, we present a framework linking competitive inhibition to alchemical free energy perturbation (FEP) calculations, focusing on Escherichia coli dihydrofolate reductase (DHFR) and its inhibition by trimethoprim (TMP). Using thermodynamic cycles, we relate experimentally measured binding constants (Ki and Km) to free energy differences associated with wild-type and mutant forms of DHFR with a mean error of 0.9 kcal/mol, providing insight into the molecular underpinnings of TMP resistance. Our findings highlight the importance of local conformational dynamics in competitive inhibition. Mutations in DHFR affect substrate and inhibitor binding affinities differently, influencing the fitness landscape under selective pressure from TMP. Our FEP simulations reveal that resistance mutations stabilize inhibitor-bound or substrate-bound states through specific structural and/or dynamical effects. The interplay of these effects showcases significant molecular-level epistasis in certain cases. The ability to separately assess substrate and inhibitor binding provides valuable insights, allowing for a more precise interpretation of mutation effects and epistatic interactions. Furthermore, we identify key challenges in FEP simulations, including convergence issues arising from charge-changing mutations and long-range allosteric effects. By integrating computational and experimental data, we provide an effective approach for predicting the functional impact of resistance mutations and their contributions to evolutionary fitness landscapes. These insights pave the way for constructing robust mutational scanning protocols and designing more effective therapeutic strategies against resistant bacterial strains.
Collapse
Affiliation(s)
| | | | - Ali Rana Atilgan
- Faculty of Engineering and
Natural Sciences, Sabanci University, 34956 Istanbul, Türkiye
| | - Canan Atilgan
- Faculty of Engineering and
Natural Sciences, Sabanci University, 34956 Istanbul, Türkiye
| |
Collapse
|
29
|
Hu Y, Chen J, Qi S, Wang H, Zhu Z, Peng Y, Wang W, Huang G, Fang Z, Ye Y, Wang Z, Guo K. Sequence Similarity Network Guided Discovery of a Dehydrogenase for Asymmetric Carbonyl Dehydrogenation. Angew Chem Int Ed Engl 2025; 64:e202501425. [PMID: 39985430 DOI: 10.1002/anie.202501425] [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: 01/17/2025] [Revised: 02/21/2025] [Accepted: 02/21/2025] [Indexed: 02/24/2025]
Abstract
Carbonyl dehydrogenation is one of the most valuable transformations in modern synthetic chemistry. As compared to traditional chemical synthesis methods, enzymatic dehydrogenation offers a greener and more selective alternative. However, except for a few rare natural dehydrogenases for desaturation, current enzymatic methods predominantly rely on enzyme promiscuity, which often suffers from lower efficiency and limited reaction controllability. Herein, we employed sequence similarity networks to mine natural dehydrogenases from a vast array of sequences with potential dehydrogenation activity. This approach led to the discovery of an uncharacterized FAD-dependent enzyme capable of efficiently performing the desymmetrizing desaturation of cyclohexanones, thereby generating diverse cyclohexenones bearing remote γ-quaternary stereocenters. The current method has enhanced the turnover frequency (TOF) by approximately 178-fold as compared to the best existing biocatalytic strategies and displayed almost no overoxidation reactions. Through a combination of experimental assays and computational studies, we elucidated that this enzyme enhances its dehydrogenation capability through an unconventional proton relay system, absent in previously reported enzyme promiscuity systems. Additionally, this streamlined enzymatic process demonstrated scalability to gram-scale synthesis with maintained efficiency and selectivity, offering robust and sustainable alternatives for the synthesis of chiral cyclohexenones with high optical purity.
Collapse
Affiliation(s)
- Yujing Hu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 211816, P. R. China
| | - Jie Chen
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 211816, P. R. China
| | - Shaofang Qi
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 211816, P. R. China
| | - Hui Wang
- Research Center for Industries of the Future, Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, Department of Chemistry, School of Science, Westlake University, Zhejiang, Hangzhou, 310024, P. R. China
| | - Zhaoxuan Zhu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agriculture University, Jiangsu, Nanjing, 210095, P. R. China
| | - Yongzhen Peng
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 211816, P. R. China
| | - Wenjing Wang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 211816, P. R. China
| | - Guixiang Huang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 211816, P. R. China
| | - Zheng Fang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 211816, P. R. China
| | - Yuxuan Ye
- Research Center for Industries of the Future, Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, Department of Chemistry, School of Science, Westlake University, Zhejiang, Hangzhou, 310024, P. R. China
| | - Zhiguo Wang
- Zhejiang Key Laboratory of Medical Epigenetics, Institute of Aging Research, School of Basic Medicine Sciences, Hangzhou Normal University, Zhejiang, Hangzhou, 311121, P. R. China
| | - Kai Guo
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 211816, P. R. China
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Jiangsu, Nanjing, 210009, P. R. China
| |
Collapse
|
30
|
Hamley IW, de Mello LR, Castelletto V, Zinn T, Cowieson N, Seitsonen J, Bizien T. Semaglutide Aggregates into Oligomeric Micelles and Short Fibrils in Aqueous Solution. Biomacromolecules 2025. [PMID: 40355389 DOI: 10.1021/acs.biomac.5c00342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Semaglutide is a lipopeptide with important applications in the treatment of diabetes, obesity, and other conditions. This class of drug (glucagon-like peptide-1 agonists and other lipidated peptides) may be susceptible to aggregation due to the tendency of lipopeptides to self-assemble into various nanostructures. Here, we show using cryogenic-TEM, small-angle X-ray scattering, and molecular dynamics simulations that semaglutide in aqueous solution undergoes slow aggregation into spherical micelles in water at sufficiently high concentration. A small population of needle-shaped fibril aggregates is also observed. At a lower concentration, dimer and trimer structures are formed. The micelles, once formed, are stable toward further aging. The aggregation influences the effect of semaglutide on the permeability of an epithelial gut model membrane of Caco-2 cells. These findings are expected to be important in understanding the long-term stability of semaglutide solutions and the potential effects of aggregation on therapeutic efficacy.
Collapse
Affiliation(s)
- Ian W Hamley
- School of Chemistry, Food Biosciences and Pharmacy, University of Reading, Whiteknights, Reading, Berkshire RG6 6AD, U.K
| | - Lucas R de Mello
- School of Chemistry, Food Biosciences and Pharmacy, University of Reading, Whiteknights, Reading, Berkshire RG6 6AD, U.K
| | - Valeria Castelletto
- School of Chemistry, Food Biosciences and Pharmacy, University of Reading, Whiteknights, Reading, Berkshire RG6 6AD, U.K
| | - Thomas Zinn
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, Oxfordshire OX11 0DE, U.K
| | - Nathan Cowieson
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, Oxfordshire OX11 0DE, U.K
| | - Jani Seitsonen
- Nanomicroscopy Center, Aalto University, Puumiehenkuja 2, FIN-02150 Espoo, Finland
| | - Thomas Bizien
- Synchrotron SOLEIL, L'Orme des Merisiers, Départementale 128, 91190 Saint-Aubin, France
| |
Collapse
|
31
|
Czyżnikowska Ż, Mysłek M, Marciniak A, Płaczek R, Kotynia A, Krzyżak E. In Silico Approach to Design of New Multi-Targeted Inhibitors Based on Quinoline Ring with Potential Anticancer Properties. Int J Mol Sci 2025; 26:4620. [PMID: 40429764 PMCID: PMC12110963 DOI: 10.3390/ijms26104620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Revised: 05/09/2025] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
Searching for new anticancer drugs is a significant challenge for the medical community due to the current limitations of existing treatments. The primary objective of this study was to design and optimize multi-targeted drug candidates based on a quinoline scaffold. In this paper, we adopt various in silico techniques, including molecular docking, molecular dynamics simulations, and ADMET property modeling, to predict the binding affinity and interactions of 7-ethyl-10-hydroxycamptothecin derivatives with multiple biological targets. The interactions of these compounds with three potential molecular targets, topoisomerase I, bromodomain-containing protein 4, and ATP-binding cassette sub-family G member 2 proteins, were analyzed. It has been previously proved that the inhibition of these molecular targets may have beneficial effects on cancer treatment. The designed chemical compounds can effectively interact with selected proteins, thereby establishing their potential as drug candidates. Molecular docking revealed promising binding affinities, with topoisomerase I docking scores ranging from -9.0 to -10.3 kcal/mol, BRD4 scores from -6.6 to -8.0 kcal/mol, and ABCG2 scores from -8.0 to -10.0 kcal/mol. Furthermore, the ADMET property analysis indicates promising pharmacological profiles, protein binding affinity, selectivity, and bioavailability while minimizing toxicity. For example, satisfactory logP values have been demonstrated in the favorable range for bioavailability after oral administration. Additionally, several compounds exhibited predicted aqueous solubility values greater than -3, suggesting moderate-to-good solubility, which is crucial for oral drug delivery.
Collapse
Affiliation(s)
- Żaneta Czyżnikowska
- Department of Basic Chemical Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland; (A.M.); (R.P.); (A.K.); (E.K.)
| | - Martyna Mysłek
- “Biomolecule” Student Science Club, Department of Basic Chemical Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland
| | - Aleksandra Marciniak
- Department of Basic Chemical Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland; (A.M.); (R.P.); (A.K.); (E.K.)
| | - Remigiusz Płaczek
- Department of Basic Chemical Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland; (A.M.); (R.P.); (A.K.); (E.K.)
| | - Aleksandra Kotynia
- Department of Basic Chemical Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland; (A.M.); (R.P.); (A.K.); (E.K.)
| | - Edward Krzyżak
- Department of Basic Chemical Sciences, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland; (A.M.); (R.P.); (A.K.); (E.K.)
| |
Collapse
|
32
|
Hu J, Islam MM, He J, Zhang L, Liu L. Tuning energy transport in helical protein nanotubes through side-chain modifications. Acta Biomater 2025:S1742-7061(25)00345-9. [PMID: 40348071 DOI: 10.1016/j.actbio.2025.05.020] [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: 01/06/2025] [Revised: 04/24/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Fibrous proteins are widely used as materials due to their biocompatibility, flexibility, and mechanical properties. With advancements in bioelectronics and flexible materials, there is increasing demand for biocompatible materials with tunable thermal conductivity. Understanding the mechanisms of thermal transport in proteins can facilitate the design of biomaterials with tailored thermal properties. In this study, we use non-equilibrium molecular dynamics (NEMD) to investigate how side-chain mass affects thermal transport in α-helix proteins. We analyze four representative residues - glycine (G), alanine (A), leucine (L), and phenylalanine (F) - and demonstrate that variations in side-chain mass significantly influence thermal conductivity. Results show that heavier side chains hinder heat transport, while lighter side chains enhance it. Phonon analysis reveals that side-chain mass primarily affects the properties of low-frequency acoustic and semi-optical phonons, which are critical for energy transfer. These findings provide insights into the design of protein-based biomaterials with customized thermal properties, offering potential applications in bioelectronics, medical devices, and sustainable materials. STATEMENT OF SIGNIFICANCE: This research explores how side chains in α-helix proteins influence their thermal conductivity through the application of molecular dynamics simulations. By analyzing four types of amino acids with differing side-chain masses, the study demonstrates that lighter side chains enhance heat transport, whereas heavier ones diminish it. This work establishes a direct correlation between protein structural features and their thermal properties, providing the groundwork that could enable the engineering of biomaterials with tailored heat conduction capabilities. The findings have implications for applications in bioelectronics, medical devices, and sustainable materials, where precise thermal management is essential, rendering this research highly relevant to scientists and engineers focused on advancing biocompatible materials with specific thermal characteristics.
Collapse
Affiliation(s)
- Jiayue Hu
- Department of Mechanical Engineering, Temple University, 1801 N Broad Street, Philadelphia, PA, 19122, USA
| | - Md Mohaiminul Islam
- Department of Mechanical Engineering, Temple University, 1801 N Broad Street, Philadelphia, PA, 19122, USA
| | - Jinlong He
- Failure Mechanics and Engineering Disaster Prevention Key Laboratory of Sichuan Province, Sichuan University, Chengdu 610207, PR China; MOE Key Laboratory of Deep Earth Science and Engineering, College of Architecture and Environment, Sichuan University, Chengdu 610065, PR China
| | - Lin Zhang
- Department of Engineering Mechanics, School of Civil Engineering, Shandong University, Jinan 250061, PR China
| | - Ling Liu
- Department of Mechanical Engineering, Temple University, 1801 N Broad Street, Philadelphia, PA, 19122, USA.
| |
Collapse
|
33
|
Liu Y, Liao B, Zhang QL. Collective Vibration Decoupling of Confined Water in Membrane Channels. J Phys Chem B 2025; 129:4432-4437. [PMID: 40271975 DOI: 10.1021/acs.jpcb.5c00172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
We previously reported that the asymmetric IR absorption of monolayer water confined within two-dimensional nanochannels is capable of nonthermally inducing a unidirectional flow [Zhang, Q. L. Phys. Rev. Lett. 2024, 132, 184003], while the reason for the difference in the collective vibration IR spectrum between the confined water (CW) and bulk water is still not fully understood. Here, using molecular dynamics simulations, we systematically demonstrated that the CW in narrow graphene membrane channels will appear as a predominant fingerprint-peak and a subpeak in the collective vibration spectrum band. A comparison with the calculated IR spectrum for the CW in the channels with different interlayer spacings revealed that the double-peaked pattern originates from the decoupling of the CW's collective vibration. The highlight spectral intensity of the fingerprint-peak is attributed to the low-cost out-of-plane vibration (wag mode) of the CW molecules. These findings help us understand the physical origins of the unique IR spectra of CW in nanochannels, thereby providing a robust theoretical support for the regulation of the CW's structure and dynamics properties by a remote terahertz stimulation.
Collapse
Affiliation(s)
- Yang Liu
- School of Mathematics-Physics and Finance, Anhui Polytechnic University, Wuhu 241000, China
| | - Bin Liao
- Key Laboratory of Mechanics, Anhui Polytechnic University, Wuhu 241000, China
| | - Qi-Lin Zhang
- School of Mathematics-Physics and Finance, Anhui Polytechnic University, Wuhu 241000, China
| |
Collapse
|
34
|
Tsujimura M, Ishikita H, Saito K. Determinants of hydrogen bond distances in proteins. Phys Chem Chem Phys 2025; 27:9794-9805. [PMID: 40275838 DOI: 10.1039/d5cp00511f] [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: 04/26/2025]
Abstract
Hydrogen bonds (H-bonds) between oxygen atoms, with the O-H bond donated to the acceptor O atom (Odonor-H⋯Oacceptor), are essential for stabilizing protein structures and facilitating enzymatic reactions. The dielectric and electrostatic environment of proteins, as well as structural constraints imposed by protein folding, influence the nature of H-bonds. In this study, we investigated how these factors affect H-bond distances in proteins. Analysis of 906 high-resolution protein structures (≤1.2 Å) from the Protein Data Bank revealed that H-bond distances for H-bonds with the same donor and acceptor groups are distributed around a value primarily determined by the pKa difference between these groups (ΔpKa) in water, with lower ΔpKa values leading to shorter distances. This correlation arises from enhanced electron redistribution from the H-bond acceptor to the donor in lower ΔpKa H-bonds, which increases the covalent character of the H-bond and decreases the H⋯Oacceptor distance. In contrast, H-bond distances are largely unaffected by whether the H-bond is buried in the protein interior or exposed to bulk water, as the strength of the electrostatic interaction between the donor and acceptor groups plays a minor role in determining distances. Furthermore, analysis of H-bonds in microbial rhodopsins using a quantum mechanical/molecular mechanical approach demonstrates that the protein environment primarily influences H-bond distances electrostatically by altering the ΔpKa of the H-bond, while structural constraints impose a secondary influence by altering Odonor-H⋯Oacceptor angles or H⋯Oacceptor distances without changing ΔpKa.
Collapse
Affiliation(s)
- Masaki Tsujimura
- Department of Advanced Interdisciplinary Studies, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8904, Japan.
| | - Hiroshi Ishikita
- Department of Applied Chemistry, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan.
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8904, Japan
| | - Keisuke Saito
- Department of Applied Chemistry, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan.
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8904, Japan
| |
Collapse
|
35
|
Chen X, Zhang Y, Li S, Liao W, Tao W, Deng Z, Bugni TS, Su H, Zhang F. Cytochrome P450 Mediated Cyclohexane Ring Formation in Forazoline Biosynthesis. Angew Chem Int Ed Engl 2025:e202504925. [PMID: 40329414 DOI: 10.1002/anie.202504925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 05/05/2025] [Accepted: 05/06/2025] [Indexed: 05/08/2025]
Abstract
Forazoline A, produced by the marine actinomycete Actinomadura sp. WMMB-499, is a unique PK/NRP hybrid macrolactone with promising antifungal in vivo efficacy through a previously unreported mechanism. Although a PKS/NRPS gene cluster was identified as a candidate for forazoline production, the precise biosynthetic pathway and the functions of the tailoring enzymes remain unclear. In this work, the functions of three cytochrome P450 mono-oxygenases (FrazP1P2P3) were characterized. Notably, FrazP2 was found to mediate cyclohexane ring formation from an 1,3,6-triene precursor during forazoline A biosynthesis, as confirmed by genetic and biochemical analysis. To gain structural and mechanistic insight into the activity of FrazP2, the crystal structure of a FrazP2-substrate complex has been solved at 2.3 Å resolution. The molecular dynamics simulations and DFT calculations revealed an unprecedented enzyme-catalyzed oxidative cyclization reaction by FrazP2. These findings expand our understanding of the catalytic diversity of cytochrome P450s, contributing to the diversification of natural products and enabling the creation of unnatural derivatives with increased antifungal potency.
Collapse
Affiliation(s)
- Xinru Chen
- Department of Pulmonary and Critical Care Medicine, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, 430071, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University, Wuhan, Hubei, 430071, China
| | - Yujie Zhang
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- University of Chinese Academy of Sciences, 19 A Yuguan Road, Beijing, 100049, China
| | - Shiqi Li
- Department of Pulmonary and Critical Care Medicine, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, 430071, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University, Wuhan, Hubei, 430071, China
| | - Weiting Liao
- Department of Pulmonary and Critical Care Medicine, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, 430071, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University, Wuhan, Hubei, 430071, China
| | - Weixin Tao
- Department of Pulmonary and Critical Care Medicine, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, 430071, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University, Wuhan, Hubei, 430071, China
| | - Zixin Deng
- Department of Pulmonary and Critical Care Medicine, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, 430071, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University, Wuhan, Hubei, 430071, China
| | - Tim S Bugni
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - Hao Su
- State Key Laboratory of Engineering Biology for Low-Carbon Manufacturing, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300308, China
- University of Chinese Academy of Sciences, 19 A Yuguan Road, Beijing, 100049, China
| | - Fan Zhang
- Department of Pulmonary and Critical Care Medicine, Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, 430071, China
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University, Wuhan, Hubei, 430071, China
| |
Collapse
|
36
|
Han L, Zhang X, Wu F, Wang T, Zhai H. Exploring the binding mode of BBA protein anchored on defective graphene and evaluating the biocompatibility of two types of graphene with λ-repressor protein. Colloids Surf B Biointerfaces 2025; 249:114510. [PMID: 39827588 DOI: 10.1016/j.colsurfb.2025.114510] [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: 10/13/2024] [Revised: 12/30/2024] [Accepted: 01/11/2025] [Indexed: 01/22/2025]
Abstract
Since defects in nanomaterials are inevitable during experimental manipulation, investigating the interactions between defective materials and active biological proteins is crucial for evaluating the biocompatibility and biosafety of nanomaterials. This study employs molecular dynamics simulation techniques to investigate the interaction mechanisms between two types of graphene (ideal graphene and defective graphene) and two model proteins (BBA protein and λ-repressor protein). The simulation results indicate that both types of graphene exhibit superior biocompatibility with the λ-repressor protein compared to the BBA protein. The difference in binding modes of the BBA protein with the two graphenes arises mainly from its initial orientation. Notably, the positively charged Arg residue forces the BBA protein to "anchor" to the surface of defective graphene, significantly restricting its lateral migration. The λ-repressor protein is "anchored" onto the surface of defective graphene through hydrogen bonding interactions involving its Ser residue. Such hydrogen bonding was never reported in similar systems. The distinctive binding modes of these two model proteins with defective graphene are beneficial for the future development of safer and more efficient nanomedicine technologies.
Collapse
Affiliation(s)
- Lu Han
- College of Chemistry & Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China
| | - Xiaoyun Zhang
- College of Chemistry & Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China.
| | - Fei Wu
- School of Nuclear Science and Technology, Lanzhou University, Lanzhou 730000, PR China
| | - Tianhua Wang
- College of Chemistry & Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China
| | - Honglin Zhai
- College of Chemistry & Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
37
|
Fernandez-Sánchez F, Flores-Ávila J, García HS, Mixcoha E, Balleza D. Molecular dynamics study of the helix-to-disorder transition in short antimicrobial peptides from Urodacus yaschenkoi. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2025; 54:135-148. [PMID: 40137971 DOI: 10.1007/s00249-025-01740-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/27/2024] [Accepted: 01/14/2025] [Indexed: 03/29/2025]
Abstract
The bioactivity of the short antimicrobial peptides (ssAMPs) UyCT1, CT2, CT3, CT5, Uy17, Uy192, and Uy234 from the scorpion Urodacus yaschenkoi has been well-characterized. The antagonistic effect reported in those studies on some clinical isolates of pathogenic bacteria, including Staphylococcus aureus, Klebsiella pneumoniae, and Escherichia coli was studied with an in silico approach to contrast their bioactivity in molecular terms. The peptides were modeled by generating high-quality structures with AlphaFold2, properly validated, and subjected to dynamic simulations in aqueous systems with the Gromos 43a1 and Charmm 36 force fields. Our analysis indicates that the degree of helicity of these peptides is closely linked to their composition and several physicochemical factors such as the hydrophobicity index, electrostatic potential, intrinsic flexibility, and dipole moment. We also found interesting parallels between the degree of order mentioned and the potency of each peptide with previously studied bacterial strains, specifically S. aureus. We analyzed in more detail of two specific peptides, UyCT1 and UyCT2, whose sequences are almost identical, except for the presence of a G-cap in the former. This subtle difference has a decisive impact on the conformational dynamics of these peptides, making the UyCT2 peptide more prone to disorder and the UyCT1 peptide more stable through the formation of multiple H-bonds. This analysis, based on an exhaustive characterization of the physicochemical properties of these ssAMPs, together with the determination of their conformational dynamics and the correlation with experimental data, could be the basis for the design and optimization of new drugs based on natural peptides found in scorpion venoms.
Collapse
Affiliation(s)
- Flora Fernandez-Sánchez
- Laboratorio de Microbiología, Unidad de Investigación y Desarrollo en Alimentos, Instituto Tecnológico de Veracruz, Tecnológico Nacional de México, Veracruz, Mexico
| | - Jenny Flores-Ávila
- Laboratorio de Microbiología, Unidad de Investigación y Desarrollo en Alimentos, Instituto Tecnológico de Veracruz, Tecnológico Nacional de México, Veracruz, Mexico
| | - Hugo S García
- Laboratorio de Microbiología, Unidad de Investigación y Desarrollo en Alimentos, Instituto Tecnológico de Veracruz, Tecnológico Nacional de México, Veracruz, Mexico
| | - Edgar Mixcoha
- Laboratorio de Microbiología, Unidad de Investigación y Desarrollo en Alimentos, Instituto Tecnológico de Veracruz, Tecnológico Nacional de México, Veracruz, Mexico
| | - Daniel Balleza
- Laboratorio de Microbiología, Unidad de Investigación y Desarrollo en Alimentos, Instituto Tecnológico de Veracruz, Tecnológico Nacional de México, Veracruz, Mexico.
| |
Collapse
|
38
|
Dey S, Field EH, Wang Y, Han K, Kumar R, Shimon LJW, Liu S, Guerin S, Berryman JT, Ji W, Reynolds NP, Bera S. Fluorination Induced Inversion of Helicity and Self-Assembly Into Cross-α Like Piezoelectric Amyloids by Minimalistic Designer Peptide. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2500288. [PMID: 40100237 DOI: 10.1002/smll.202500288] [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: 01/08/2025] [Revised: 02/22/2025] [Indexed: 03/20/2025]
Abstract
Although initially identified as pathological aggregates, amyloid fibrillar assemblies formed by various proteins and peptides are now known to have crucial physiological roles, carrying out numerous biological functions in almost all organisms. Due to unique features, the common etiology of amyloids' cross-β structure is long posited as a template for designing artificial self-assembling systems. However, the recent discovery of cross-α amyloids indicates additional structural paradigms for self-assembly into ordered nanostructures, turning significant attention toward designing artificial nanostructures based on cross-α assembly. Herein, a minimalistic designer peptide which forms a hydrogen-bonded amyloid-like structure while remaining in the α region of conformation is engineered, to investigate the effect of aromatic, hydrophobic, and steric considerations on amyloidal assemblies. These results demonstrate a significant modulation of helicity and self-assembly, leading to the structure-dependent piezoelectric function of the amyloid-like cross-α fibrils. This study indicates a potential avenue for molecular engineering of functional peptide materials.
Collapse
Affiliation(s)
- Sukantha Dey
- Department of Chemistry, Ashoka University, Sonipat, Haryana, 131029, India
| | - Emily H Field
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Yuehui Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Kyunghoon Han
- Department of Physics and Materials Science, University of Luxembourg, Luxembourg City, L-1511, Luxembourg
| | - Rohit Kumar
- Department of Chemistry, Ashoka University, Sonipat, Haryana, 131029, India
| | - Linda J W Shimon
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Shuaijie Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Sarah Guerin
- Department of Chemical Sciences, Bernal Institute, University of Limerick, Limerick, V94 T9PX, Ireland
- SSPC, The Science Foundation Ireland Research Centre for Pharmaceuticals, University of Limerick, Limerick, V94 T9PX, Ireland
| | - Joshua T Berryman
- Department of Physics and Materials Science, University of Luxembourg, Luxembourg City, L-1511, Luxembourg
| | - Wei Ji
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Nicholas P Reynolds
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, Victoria, 3086, Australia
- The Biomedical and Environmental Sensor Technology (BEST) Research Centre, Biosensors Program, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, Victoria, 3086, Australia
| | - Santu Bera
- Department of Chemistry, Ashoka University, Sonipat, Haryana, 131029, India
| |
Collapse
|
39
|
Raghuraman P, Park S. Exploring the modulation of phosphorylation and SUMOylation-dependent NPR1 conformational switching on immune regulators TGA3 and WRKY70 through molecular simulation. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2025; 222:109711. [PMID: 40056739 DOI: 10.1016/j.plaphy.2025.109711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/12/2025] [Accepted: 02/24/2025] [Indexed: 03/10/2025]
Abstract
NPR1 (Nonexpressor pathogenesis-related genes 1) is regulated by multisite phosphorylation and SUMOylation, serving as a master switch for effector-triggered plant immunity through a transcriptional activator (TGA3) and repressor (WRKY70) are experimentally well studied. However, the conformational relationship between the various phosphorylation, un-phosphorylation states, and SUMOylation's role in the functional switch remains unclear. Using deep learning-based molecular modeling, docking, and multi-nanosecond simulations (totaling 2 μs) with end-state free energy calculations, we unveil how different phosphorylation states impact the dynamic stability of NPR1's four phospho-serine residues (Ser11, Ser15, Ser55, & Ser59) and binding of the TGA3-WRKY70 over SUMOylation. Results from our simulations show that the salicylic-acid induced P-Ser11/15NPR1-SUMO3 stabilizes helices and the flexible activation loop (α22Lys423 - α1Arg50 & L35Asp467-Arg51α51, and Gly27L3), thereby switching association with TGA3. The inter-helix salt-bridge formed (L10Arg99-Glu323α9 and α14Glu280-Pro264L6) between the phosphorylated NPR1-SUMO3-TGA3 engage in tight control of conformational regulation were disengaged in the unphosphorylated system. The P-Ser55/59NPR1-SUMO3-WRKY70 reorients itself and forms an electrostatic and hydrogen bond with Lys145α7 - L2Asp26, L6Arg99 - Leu293L18 and Lys262L15 - Glu241L15, α13Val239 (α310), & L17Leu267 keeps complex stable and quiescent compare to unphosphorylated NPR1-WRKY70. Subsequently, the essential dynamic and secondary structural analysis reveals that the phosphorylation inhibits the α516 (long helix) formation and reduces the communication space between the 460α25-βturn3-α30-L42590 (NPR1) and 90L9-L10107 (SUMO3), making the binding more suitable for TGA3 (260βturn-L6270) and WRKY70 (230L15-L16265) via activation loop. These findings, which reveal the atomic and structural details of the NPR1's post-translational modification, will illuminate future investigations into enhancing plant immunity.
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
|
40
|
Niu H, Li MY, Gao Y, Li JG, Jiang J, Ying YL, Long YT. Direct mapping of tyrosine sulfation states in native peptides by nanopore. Nat Chem Biol 2025; 21:716-726. [PMID: 39322788 DOI: 10.1038/s41589-024-01734-x] [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: 11/08/2023] [Accepted: 08/19/2024] [Indexed: 09/27/2024]
Abstract
Sulfation is considered the most prevalent post-translational modification (PTM) on tyrosine; however, its importance is frequently undervalued due to difficulties in direct and unambiguous determination from phosphorylation. Here we present a sequence-independent strategy to directly map and quantify the tyrosine sulfation states in universal native peptides using an engineered protein nanopore. Molecular dynamics simulations and nanopore mutations reveal specific interactions between tyrosine sulfation and the engineered nanopore, dominating identification across diverse peptide sequences. We show a nanopore framework to discover tyrosine sulfation in unknown peptide fragments digested from a native protein and determine the sequence of the sulfated fragment based on current blockade enhancement induced by sulfation. Moreover, our method allows direct observation of peptide sulfation in ultra-low abundance, down to 1%, and distinguishes it from isobaric phosphorylation. This sequence-independent strategy suggests the potential of nanopore to explore specific PTMs in real-life samples and at the omics level.
Collapse
Affiliation(s)
- Hongyan Niu
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, China
| | - Meng-Yin Li
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China.
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, China.
| | - Yan Gao
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
| | - Jun-Ge Li
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
| | - Jie Jiang
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
| | - Yi-Lun Ying
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, China
| | - Yi-Tao Long
- Molecular Sensing and Imaging Center, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, China.
| |
Collapse
|
41
|
Sarma S, Dowerah D, Basumatary M, Phonglo A, Deka RC. Inhibitory potential of furanocoumarins against cyclin dependent kinase 4 using integrated docking, molecular dynamics and ONIOM methods. J Biomol Struct Dyn 2025; 43:3974-4003. [PMID: 38189343 DOI: 10.1080/07391102.2023.2300755] [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: 10/04/2023] [Accepted: 12/23/2023] [Indexed: 01/09/2024]
Abstract
Cyclin Dependent Kinase 4 (CDK4) is vital in the process of cell-cycle and serves as a G1 phase checkpoint in cell division. Selective antagonists of CDK4 which are in use as clinical chemotherapeutics cause various side-effects in patients. Furanocoumarins induce anti-cancerous effects in a range of human tumours. Therefore, targeting these compounds against CDK4 is anticipated to enhance therapeutic effectiveness. This work intended to explore the CDK4 inhibitory potential of 50 furanocoumarin molecules, using a comprehensive approach that integrates the processes of docking, drug-likeness, pharmacokinetic analysis, molecular dynamics simulations and ONIOM (Our own N-layered Integrated molecular Orbital and Molecular mechanics) methods. The top five best docked compounds obtained from docking studies were screened for subsequent analysis. The molecules displayed good pharmacokinetic properties and no toxicity. Epoxybergamottin, dihydroxybergamottin and notopterol were found to inhabit the ATP-binding zone of CDK4 with substantial stability and negative binding free energy forming hydrogen bonds with key catalytic residues of the protein. Notopterol exhibiting the highest binding energy was subjected to ONIOM calculations wherein the hydrogen bonding interactions were retained with significant negative interaction energy. Hence, through these series of computerised methods, notopterol was screened as a potent CDK4 inhibitor and can act as a starting point in successive processes of drug design.
Collapse
Affiliation(s)
- Srutishree Sarma
- CMML-Catalysis and Molecular Modelling Lab, Department of Chemical Sciences, Tezpur University, Sonitpur, Assam, India
| | - Dikshita Dowerah
- CMML-Catalysis and Molecular Modelling Lab, Department of Chemical Sciences, Tezpur University, Sonitpur, Assam, India
| | - Moumita Basumatary
- CMML-Catalysis and Molecular Modelling Lab, Department of Chemical Sciences, Tezpur University, Sonitpur, Assam, India
| | - Ambalika Phonglo
- CMML-Catalysis and Molecular Modelling Lab, Department of Chemical Sciences, Tezpur University, Sonitpur, Assam, India
| | - Ramesh Ch Deka
- CMML-Catalysis and Molecular Modelling Lab, Department of Chemical Sciences, Tezpur University, Sonitpur, Assam, India
| |
Collapse
|
42
|
Bellucci L, Capone M, Daidone I, Zanetti-Polzi L. Conformational heterogeneity and protonation equilibria shape the photocycle branching in channelrhodopsin-2. Int J Biol Macromol 2025; 305:140977. [PMID: 39956237 DOI: 10.1016/j.ijbiomac.2025.140977] [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: 12/02/2024] [Revised: 01/24/2025] [Accepted: 02/11/2025] [Indexed: 02/18/2025]
Abstract
Channelrhodopsin-2 is a photoactive membrane protein serving as an ion channel, gathering significant interest for its applications in optogenetics. Despite extensive investigation, several aspects of its photocycle remain elusive and continue to be subjects of ongoing debate. Of particular interest are the localization of the P480 intermediate within the photocycle and the timing of the deprotonation of glutamic acid E90, a critical residue for ChR2 functioning. In this study, we explore the possibility of an early-P480 state, formed directly upon photoillumination of the dark-adapted state, where E90 is deprotonated, as hypothesized in a previous work [Kuhne et al. Proc. Natl. Acad. Sci. 116.19 (2019): 9380]. Employing extended molecular dynamics simulations, deprotonation free energy calculations, and the computation of the infrared band associated with E90, we provide support to the photocycle model proposed by Kuhne et al. Furthermore, our findings show that E90 protonation state is influenced by diverse interconnected variables and provide molecular detail insights that connect E90 interaction pattern with its deprotonation propensity. Our data demonstrate in fact that both protonated and deprotonated E90 are possible in P480 depending on E90 hydrogen bonding pattern and explaining the molecular mechanism at the basis of P480 accumulation under continuous illumination.
Collapse
Affiliation(s)
- Luca Bellucci
- NEST-SNS, CNR Institute of Nanoscience, Piazza San Silvestro 12, Pisa 5612, Italy
| | - Matteo Capone
- Center S3, CNR Institute of Nanoscience, Via Campi 213/A, Modena 41125, Italy
| | - Isabella Daidone
- Department of Physical and Chemical Sciences, University of L'Aquila, via Vetoio (Coppito 1), L'Aquila 67010, Italy
| | - Laura Zanetti-Polzi
- Center S3, CNR Institute of Nanoscience, Via Campi 213/A, Modena 41125, Italy.
| |
Collapse
|
43
|
Sardag I, Duvenci ZS, Belkaya S, Timucin E. Computational modeling of the anti-inflammatory complexes of IL37. J Mol Graph Model 2025; 136:108952. [PMID: 39854883 DOI: 10.1016/j.jmgm.2025.108952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/31/2024] [Accepted: 01/11/2025] [Indexed: 01/27/2025]
Abstract
Interleukin (IL) 37 is an anti-inflammatory cytokine belonging to the IL1 protein family. Owing to its pivotal role in modulating immune responses, elucidating the IL37 complex structures holds substantial therapeutic promise for various autoimmune disorders and cancers. However, none of the structures of IL37 complexes have been experimentally characterized. This computational study aims to address this gap through molecular modeling and classical molecular dynamics simulations. We modeled all protein-protein complexes of IL37 using a range of methods from homology modeling to AlphaFold2 multimer predictions. Models that successfully recapitulated experimental features underwent further analysis through molecular dynamics simulations. As positive controls, binary and ternary complexes of IL18 from PDB were included for comparison. Several key findings emerged from the comparative analysis of IL37 and IL18 complexes. IL37 complexes exhibited higher mobility than the IL18 complexes. Simulations of the IL37-IL18Rα complex revealed altered receptor conformations capable of accommodating a dimeric IL37, with the N-terminal loop of IL37 contributing significantly to complex mobility. Additionally, the glycosyl chain on N297 of IL18Rα, which contours one edge of the cytokine binding surface, acted as a steric block against the N-terminal loop of IL37. Further, investigations into interactions between IL37 and IL18BP suggested that a binding mode homologous to IL18 was unstable for IL37, indicating an alternative binding mechanism. Altogether, this study accesses to the structure and dynamics of IL37 complexes, revealing the structural underpinnings of the IL37's modulatory effect on the IL18 signaling pathway.
Collapse
Affiliation(s)
- Inci Sardag
- Bogazici University, Department of Molecular Biology and Genetics, Istanbul 34342, Turkey
| | - Zeynep Sevval Duvenci
- Acibadem University, Institute of Health Sciences Department of Biostatistics and Bioinformatics, Istanbul 34752, Turkey
| | - Serkan Belkaya
- Bilkent University, Department of Molecular Biology and Genetics, Ankara 06800, Turkey
| | - Emel Timucin
- Acibadem University, Institute of Health Sciences Department of Biostatistics and Bioinformatics, Istanbul 34752, Turkey; Acibadem University, School of Medicine Biostatistics and Medical Informatics, Istanbul 34752, Turkey.
| |
Collapse
|
44
|
Chang XP, Fan FR, Liu K, Lv HT, Zhao G, Zhang TS, Xie BB. Quantum Mechanics/Molecular Mechanics Studies on the Excited-State Relaxation Mechanisms of Cytidine Analogues: 2'-Deoxy-5-Methylcytidine and 2'-Deoxy-5-Hydroxymethylcytidine in Aqueous Solution. J Phys Chem A 2025; 129:3817-3828. [PMID: 40257932 DOI: 10.1021/acs.jpca.4c08520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
We have used the high-level QM(CASPT2//CASSCF)/MM method to investigate the excited-state properties and decay pathways of two important cytidine analogues, i.e., 2'-deoxy-5-methylcytidine (5mdCyd) and 2'-deoxy-5-hydroxymethylcytidine (5hmdCyd), in aqueous solution. In view of the computed minimum-energy structures, conical intersections, and crossing points, and the relevant excited-state decay paths including the different internal conversion (IC) and intersystem crossing (ISC) routes in and between the S1, T1, T2, and S0 states, we finally provided the feasible excited-state relaxation mechanisms of these two important epigenetic DNA nucleosides. Upon 285 nm photoexcitation, the lowest spectroscopically bright S1(ππ*) state is initially populated in the Franck-Condon (FC) region in both solvated systems and then mainly occurs direct IC to the ground state through the nearby accessible S1/S0 conical intersection, with the QM(CASPT2)/MM computed energy barriers of 9.5 and 1.6 kcal/mol for 5mdCyd and 5hmdCyd, respectively. In addition, the S1(ππ*) state can partially hop to the T1(ππ*) state directly or is mediated by the T2(ππ*) state. In comparison to the favorable singlet-mediated IC channel, the minor S1→T1 and S1→T2→T1 ISCs would take place slowly. Subsequently, the T1 state will further approach the nearby T1/S0 crossing point to slowly deactivate to the S0 state. Due to the T1/S0 crossing point above the T1-MIN as well with the small T1/S0 SOC, i.e., 9.8 kcal/mol and 0.3 cm-1 in 5mdCyd and 8.7 kcal/mol and 1.9 cm-1 in 5hmdCyd, the slow ISC would trap the system in the T1 state for a long time. The present work rationalizes the excited-state dynamics of 5mdCyd and 5hmdCyd in aqueous solution and could provide mechanistic insights into understanding the photophysics and photochemistry of similar epigenetic DNA nucleosides and their derivatives.
Collapse
Affiliation(s)
- Xue-Ping Chang
- College of Chemistry and Chemical Engineering, Green Catalysis & Synthesis Key Laboratory of Xinyang City, Xinyang Normal University, Xinyang 464000, P. R. China
| | - Feng-Ran Fan
- College of Chemistry and Chemical Engineering, Green Catalysis & Synthesis Key Laboratory of Xinyang City, Xinyang Normal University, Xinyang 464000, P. R. China
| | - Ke Liu
- College of Chemistry and Chemical Engineering, Green Catalysis & Synthesis Key Laboratory of Xinyang City, Xinyang Normal University, Xinyang 464000, P. R. China
| | - Hai-Ting Lv
- College of Chemistry and Chemical Engineering, Green Catalysis & Synthesis Key Laboratory of Xinyang City, Xinyang Normal University, Xinyang 464000, P. R. China
| | - Geng Zhao
- College of Chemistry and Chemical Engineering, Green Catalysis & Synthesis Key Laboratory of Xinyang City, Xinyang Normal University, Xinyang 464000, P. R. China
| | - Teng-Shuo Zhang
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, P. R. China
| | - Bin-Bin Xie
- Hangzhou Institute of Advanced Studies, Zhejiang Normal University, Hangzhou 311231, P. R. China
| |
Collapse
|
45
|
Hacisuleyman A, Gul A, Erman B. Role of Mutual Information Profile Shifts in Assessing the Pathogenicity of Mutations on Protein Functions: The Case of Pyrin Variants Associated With Familial Mediterranean Fever. Proteins 2025; 93:1035-1053. [PMID: 39739522 DOI: 10.1002/prot.26795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/02/2025]
Abstract
This study presents a novel method to assess the pathogenicity of pyrin protein mutations by using mutual information (MI) as a measure to quantify the correlation between residue motions or fluctuations and associated changes affecting the phenotype. The concept of MI profile shift is presented to quantify changes in MI upon mutation, revealing insights into residue-residue interactions at critical positions. We apply this method to the pyrin protein variants, which are associated with an autosomal recessively inherited disease called familial Mediterranean fever (FMF) since the available tools do not help predict the pathogenicity of the most penetrant variants. We demonstrate the utility of MI profile shifts in assessing the effects of mutations on protein stability, function, and disease phenotype. The importance of MI shifts, particularly the negative shifts observed in the pyrin example, as indicators of severe functional effects is emphasized. Additionally, the exploration of potential compensatory mechanisms suggested by positive MI shifts, which are otherwise random and inconsequential, is highlighted. The study also discusses challenges in relating MI profile changes to disease severity and advocates for comprehensive analysis considering genetic, environmental, and stochastic factors. Overall, this study provides insights into the molecular mechanisms underlying the pathogenesis of FMF and offers a framework for identifying potential therapeutic targets based on MI profile changes induced by mutations.
Collapse
Affiliation(s)
- Aysima Hacisuleyman
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Chemical and Biological Engineering, Koc University, Istanbul, Turkey
| | - Ahmet Gul
- Division of Rheumatology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Burak Erman
- Chemical and Biological Engineering, Koc University, Istanbul, Turkey
| |
Collapse
|
46
|
Asthagiri DN, Valiya Parambathu A, Beck TL. Consequences of the failure of equipartition for the p- V behavior of liquid water and the hydration free energy components of a small protein. Chem Sci 2025; 16:7503-7512. [PMID: 40160355 PMCID: PMC11951161 DOI: 10.1039/d4sc08437c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Earlier we showed that in the molecular dynamics simulation of a rigid model of water it is necessary to use an integration time-step δ t ≤ 0.5 fs to ensure equipartition between translational and rotational modes. Here we extend that study in the NVT ensemble to NpT conditions and to an aqueous protein. We study neat liquid water with the rigid, SPC/E model and the protein BBA (PDB ID: 1FME) solvated in the rigid, TIP3P model. We examine integration time-steps ranging from 0.5 fs to 4.0 fs for various thermostat plus barostat combinations. We find that a small δ t is necessary to ensure consistent prediction of the simulation volume. Hydrogen mass repartitioning alleviates the problem somewhat, but is ineffective for the typical time-step used with this approach. The compressibility, a measure of volume fluctuations, and the dielectric constant, a measure of dipole moment fluctuations, are also seen to be sensitive to δ t. Using the mean volume estimated from the NpT simulation, we examine the electrostatic and van der Waals contribution to the hydration free energy of the protein in the NVT ensemble. These contributions are also sensitive to δ t. In going from δ t = 2 fs to δ t = 0.5 fs, the change in the net electrostatic plus van der Waals contribution to the hydration of BBA is already in excess of the folding free energy reported for this protein.
Collapse
Affiliation(s)
| | | | - Thomas L Beck
- Oak Ridge National Laboratory One Bethel Valley Road Oak Ridge TN 37830 USA
| |
Collapse
|
47
|
Fang G, Dong Q, Shen X, Ye R, Chang Y, Pu K, Tao Y, Gao X, Zhou R, Ge C. Modulation of Bacterial Iron Homeostasis to Enhance Cuproptosis-like Death for the Treatment of Infected Diabetic Wound. ACS NANO 2025; 19:15578-15595. [PMID: 40252040 DOI: 10.1021/acsnano.4c17071] [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: 04/21/2025]
Abstract
Cuproptosis, an emerging cell death pathway, offers an alternative approach for antimicrobial therapy, but it suffers from deficiencies and health risks. Here, we design hollow Cu-enriched Prussian blue-based nanostructures (Cu-HMPBs) and find that the infected microenvironment facilitates the release of Cu ions from Cu-HMPBs, leading to Cu overload in bacterial cells. Meanwhile, Fe ions in bacterial cells are highly selectively chelated, triggering iron starvation. As a result, the proteotoxic stress and redox imbalance induced by Cu overload are aggravated upon iron starvation, thus remarkably enhancing cuproptosis-like bacterial cell death at extremely low-dose (noncytotoxic) Cu ions. Moreover, we demonstrate the effectiveness of this iron starvation-augmented antimicrobial strategy, and its efficacy is further validated in a methicillin-resistant Staphylococcus aureus (MRSA)-infected diabetic mouse wound model. Collectively, these findings provide a promising and universal strategy on iron starvation sensitizing cuproptosis-like bacterial cell death for combating drug resistance.
Collapse
Affiliation(s)
- Ge Fang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Qingrong Dong
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
- College of Medical Imaging, Shanxi Medical University, Taiyuan 030001, China
| | - Xiaomei Shen
- College of chemistry and materials, Jiangxi Normal University, Nanchang 330022, China
| | - Rui Ye
- School of Physics, Institute of Quantitative Biology, Zhejiang University, Hangzhou 310058, China
- Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Groningen 9747 AG, The Netherlands
| | - Yuchen Chang
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, The University of Sydney, Sydney, NSW 2006, Australia
| | - Kefeng Pu
- Nano-Bio-Chem Centre, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yujie Tao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Xingfa Gao
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Ruhong Zhou
- MOE Key Laboratory of Biosystems Homeostasis & Protection, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
- School of Physics, Institute of Quantitative Biology, Zhejiang University, Hangzhou 310058, China
| | - Cuicui Ge
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| |
Collapse
|
48
|
Suryabrahmam B, Chodnicki P, Sappati S, Jurkowski M, Agrawal A, Czub J, Raghunathan VA. Influence of Alcohols on the Bending Rigidity and the Thickness of Phospholipid Membranes: The Role of Chain Length Mismatch. J Phys Chem B 2025; 129:3988-3997. [PMID: 40233353 DOI: 10.1021/acs.jpcb.4c04282] [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
Alcohols influence the shape of the cells. To elucidate this phenomenon and understand the influence of alcohols on the mechanical properties of cell membranes such as bending rigidity, it is essential to investigate their effects on lipid bilayers. In this study, we explored the impact of short- and medium-chain alcohols on the bending rigidity and thickness of 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) bilayers in the fluid phase. We employed various techniques, including vesicle fluctuation analysis, small-angle X-ray scattering, and differential scanning calorimetry. Experimental observations were further validated and interpreted using atomistic molecular dynamics simulations. Our results demonstrate that alcohols ranging from ethanol to octanol reduce the main phase transition temperature (Tm), bending rigidity (κ), and thickness of the bilayer (DHH). Decanol and dodecanol, on the other hand, increase Tm without significantly affecting κ and the bilayer thickness. Our study conclusively shows that alcohols shorter than decanol induce a negative chain length mismatch condition, leading to disorder and enhanced interdigitation in DMPC membranes, resulting in membrane thinning and softening. In contrast, decanol, whose chain length matches that of the lipid, enhances lipid chain order and reduces their interdigitation, resulting in no alteration in the bending rigidity and membrane thickness.
Collapse
Affiliation(s)
- Buti Suryabrahmam
- Raman Research Institute, C.V. Raman Avenue, Sadashivnagar, Bengaluru 560080, India
| | - Pawel Chodnicki
- Department of Physical Chemistry, Gdańsk University of Technology, Narutowicza St 11/12, Gdańsk 80-233, Poland
| | - Subrahmanyam Sappati
- Raman Research Institute, C.V. Raman Avenue, Sadashivnagar, Bengaluru 560080, India
- Department of Physical Chemistry, Gdańsk University of Technology, Narutowicza St 11/12, Gdańsk 80-233, Poland
- Department of Pharmaceutical Technology and Biochemistry, Gdańsk University of Technology, Narutowicza St 11/12, Gdańsk 80-233, Poland
- BioTechMed Center, Gdańsk University of Technology, Narutowicza St 11/12, Gdańsk 80-233, Poland
| | - Michal Jurkowski
- Department of Physical Chemistry, Gdańsk University of Technology, Narutowicza St 11/12, Gdańsk 80-233, Poland
| | - Ayush Agrawal
- Raman Research Institute, C.V. Raman Avenue, Sadashivnagar, Bengaluru 560080, India
| | - Jacek Czub
- Department of Physical Chemistry, Gdańsk University of Technology, Narutowicza St 11/12, Gdańsk 80-233, Poland
- BioTechMed Center, Gdańsk University of Technology, Narutowicza St 11/12, Gdańsk 80-233, Poland
| | - V A Raghunathan
- Raman Research Institute, C.V. Raman Avenue, Sadashivnagar, Bengaluru 560080, India
| |
Collapse
|
49
|
Pham NT, Le HG, Peng BR, Chen LY, El-Shazly M, Su JH, Lee MH, Lai KH. Computational analysis of zoanthamine alkaloids from Zoanthus sp. as potential DKK1 and GSK-3β inhibitors for osteoporosis therapy via Wnt signaling. Sci Rep 2025; 15:14297. [PMID: 40274944 PMCID: PMC12022124 DOI: 10.1038/s41598-025-97537-8] [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: 12/25/2024] [Accepted: 04/04/2025] [Indexed: 04/26/2025] Open
Abstract
Marine invertebrates are a rich source of structurally diverse secondary metabolites with broad biological activities, making them valuable for drug discovery. The genus Zoanthus is particularly noteworthy, producing numerous bioactive alkaloids, including the zoanthamines, which show promise in treating osteoporosis. Osteoporosis, a debilitating bone disease characterized by reduced bone mineral density and increased fracture risk, is linked to Wnt signaling pathway dysregulation. This highly conserved pathway maintains tissue homeostasis and is crucial for neurogenesis, synapse formation, and bone development. Dickkopf-1 (DKK1) and glycogen synthase kinase-3β (GSK-3β), key Wnt pathway regulators, are established therapeutic targets for osteoporosis. This study employed an integrated computational approach-combining molecular docking, extensive molecular dynamics (MD) simulations, and density functional theory (DFT) calculations-to assess the inhibitory potential of 69 zoanthamine-type alkaloids against DKK1 and GSK-3β. MD simulations, analyzing root mean square deviation (RMSD), root mean square fluctuation (RMSF), radius of gyration, and free energy landscape, provided insights into protein-ligand complex stability and key interactions. Binding free energies were calculated using the MM-PBSA method combined with interaction entropy. DFT calculations further elucidated the electronic structure and reactivity of the most promising inhibitors (3α-hydroxyzoanthenamine, epioxyzoanthamine, 7α-hydroxykuroshine E, and norzoanthamine), which exhibited favorable binding interactions with key residues in target proteins. This integrative approach demonstrates the power of computational methods in drug discovery, highlighting the potential of zoanthamine alkaloids as lead compounds for innovative osteoporosis therapies.
Collapse
Affiliation(s)
- Ngoc-Thac Pham
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, 110301, Taiwan
| | - Huong-Giang Le
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, 110301, Taiwan
| | - Bo-Rong Peng
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, 110301, Taiwan
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, 333324, Taiwan
| | - Lo-Yun Chen
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, 110301, Taiwan
| | - Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Organization of African Unity Street, Abassia, Cairo, 11566, Egypt
| | - Jui-Hsin Su
- National Museum of Marine Biology and Aquarium, Pingtung, 944401, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, 804201, Taiwan
| | - Mei-Hsien Lee
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, 110301, Taiwan
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, 110301, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, 110301, Taiwan
| | - Kuei-Hung Lai
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, 110301, Taiwan.
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, 110301, Taiwan.
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, 110301, Taiwan.
| |
Collapse
|
50
|
Hasse T, Zhang Z, Huang YMM. In Silico Discovery of Potential Inhibitors Targeting the MEIG1-PACRG Complex for Male Contraceptive Development. Proteins 2025. [PMID: 40265567 DOI: 10.1002/prot.26829] [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: 12/13/2024] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/24/2025]
Abstract
The interaction between meiosis-expressed gene 1 (MEIG1) and Parkin co-regulated gene (PACRG) is a critical determinant of spermiogenesis, the process by which round spermatids mature into functional spermatozoa. Disruption of the MEIG1-PACRG complex can impair sperm development, highlighting its potential as a therapeutic target for addressing male infertility or for the development of non-hormonal contraceptive methods. This study used virtual screening, molecular docking, and molecular dynamics (MD) simulations to identify small molecule inhibitors targeting the MEIG1-PACRG interface. MD simulations provided representative protein conformations, which were used to virtually screen a library of 821 438 compounds, resulting in 48 high-ranking candidates for each protein. PACRG emerged as a favorable target due to its flexible binding pockets and better docking scores compared to MEIG1. Key binding residues with compounds included W50, Y68, N70, and E74 on MEIG1, and K93, W96, E101, and H137 on PACRG. MD simulations revealed that compound stability in MEIG1 complexes is primarily maintained by hydrogen bonding with E74 and π-π stacking interactions with W50 and Y68. In PACRG complexes, compound stabilization is facilitated by hydrogen bonding with E101 and π-π interactions involving W96 and H137. These findings highlight distinct molecular determinants of ligand binding for each protein. Our work provides mechanistic insights and identifies promising compounds for further experimental validation, establishing a foundation for developing MEIG1-PACRG interaction inhibitors as male contraceptives.
Collapse
Affiliation(s)
- Timothy Hasse
- Department of Physics and Astronomy, Wayne State University, Detroit, Michigan, USA
| | - Zhibing Zhang
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Yu-Ming M Huang
- Department of Physics and Astronomy, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|