Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (418)

Search Parameters:
Keywords = conformational mutant

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 2563 KB  
Article
Structural and Catalytic Roles of the Disulfide Bonds Cys19–Cys154 and Cys134–Cys199 in Trypsin-like Proteases: Evolutionary Insights for Disulfide Bond Acquisition
by Maiko Minakata, Yuri Murakami, Orika Ashida, Miki Matsuzaki, Kairi Ogawa, Nanako Saeki, Shigeru Shimamoto, Mitsuhiro Miyazawa, Yuji Hidaka and Nana Sakata
Molecules 2026, 31(2), 351; https://doi.org/10.3390/molecules31020351 - 19 Jan 2026
Viewed by 159
Abstract
Trypsin is one of the most extensively studied enzymes in biochemistry. However, little information is available on the role of the disulfide bonds to establish the correct conformation and enzyme activity during molecular evolution. To obtain this information, two additional disulfide bonds corresponding [...] Read more.
Trypsin is one of the most extensively studied enzymes in biochemistry. However, little information is available on the role of the disulfide bonds to establish the correct conformation and enzyme activity during molecular evolution. To obtain this information, two additional disulfide bonds corresponding to those found in human trypsin were individually or simultaneously introduced into the trypsin-like protease cocoonase (Bombyx mori), which contains three consensus disulfide bonds, and structural effects were analyzed. Enzyme assays of the mutant proteins revealed that, during molecular evolution, the Cys19–Cys154 bond contributed to improving substrate recognition (Km), whereas the Cys134–Cys199 bond contributed to enhancing catalytic turnover (kcat). In addition, the Cys134–Cys199 disulfide bond significantly increased the structural stability, whereas the Cys19–Cys154 disulfide bond promoted a more compact folded ensemble. Interestingly, when both disulfide bridges were introduced together, their effects acted synergistically, yielding the highest catalytic activity toward the substrate BAEE (kcat/Km). Taken together, these findings suggest that trypsin-like proteases evolved through a two-step adaptive process: an initial phase in which the catalytic efficiency (kcat) and structural stability were enhanced, followed by a second phase in which the fold became more compact, thereby improving the overall enzymatic activity. Full article
(This article belongs to the Special Issue Peptide and Protein Folding)
Show Figures

Graphical abstract

24 pages, 4677 KB  
Article
Functional Analysis of CRISPR-Cas9-Mediated Gene Deletion in E. coli DH5α on Membrane Permeability and Transformation Efficiency
by Feifan Leng, Xinyi Liu, Jinli He, Yubo Wang, Ning Zhu, Xiaopeng Guo, Wen Luo and Yonggang Wang
Microorganisms 2026, 14(1), 198; https://doi.org/10.3390/microorganisms14010198 - 15 Jan 2026
Viewed by 315
Abstract
This research utilized the CRISPR/Cas9 editing method to generate six mutant strains of Escherichia coli (E. coli) DH5α targeting specific genes. The functional characterization and phenotypic analysis confirmed the regulatory roles of these genes in modifying membrane permeability. The variations in [...] Read more.
This research utilized the CRISPR/Cas9 editing method to generate six mutant strains of Escherichia coli (E. coli) DH5α targeting specific genes. The functional characterization and phenotypic analysis confirmed the regulatory roles of these genes in modifying membrane permeability. The variations in membrane permeability among the mutant strains were assessed by measuring electrical conductivity, ortho-nitrophenyl-β-D-galactopyranoside (ONPG) hydrolysis, and propidium iodide (PI) fluorescence, with E. coli DH5α:ompA′ exhibiting the most pronounced increase in membrane permeability. The function of these genes in transformation was analyzed from physicochemical and microscopic perspectives. Assays of plasmid transformation efficiency revealed a significant enhancement in the E. coli DH5α:ompA′ mutant strain, underscoring the critical function of outer membrane proteins in DNA acquisition. Permeability simulations were performed utilizing the E. coli DH5α:ompA′ mutant strain, grounded in a previously established model. The quantitative correlation between transformation efficiency and membrane permeability in this mutant conformed to the equation T = aP + c. Full article
(This article belongs to the Section Environmental Microbiology)
Show Figures

Figure 1

20 pages, 7145 KB  
Article
The Role of Active Site Hydrophobic Interactions in Facilitating Catalysis in Human Terminal Deoxynucleotidyl Transferase
by Svetlana I. Senchurova, Timofey E. Tyugashev and Nikita A. Kuznetsov
Int. J. Mol. Sci. 2026, 27(1), 178; https://doi.org/10.3390/ijms27010178 - 23 Dec 2025
Viewed by 361
Abstract
Terminal deoxynucleotidyl transferase (TdT) is a unique DNA polymerase that catalyzes template-independent nucleotide addition at the 3′-end of DNA, playing a critical role in generating immune receptor diversity. While the structural importance of Loop1 in blocking template strand binding and enabling this activity [...] Read more.
Terminal deoxynucleotidyl transferase (TdT) is a unique DNA polymerase that catalyzes template-independent nucleotide addition at the 3′-end of DNA, playing a critical role in generating immune receptor diversity. While the structural importance of Loop1 in blocking template strand binding and enabling this activity is established, the precise molecular contribution of hydrophobic interactions within Loop1 to the catalytic mechanism of human TdT remains unclear. In the present study, we aim to elucidate the roles of hydrophobic Loop1 residues (L397, F400, F404) in the structural organization and catalytic function of TdT. We engineered alanine and tryptophan substitutions at these positions and systematically analyzed the resulting mutant forms using molecular dynamics simulations and pre-steady-state kinetic measurements. Our results show that substitutions L397A and F400A increase Loop1 flexibility and significantly reduce catalytic activity, particularly for purine nucleotide incorporation, while F404A completely abolishes enzymatic function. The F404W mutant largely preserves activity. All mutant forms retain the ability to bind single-stranded DNA and dNTP, but in some cases, their affinity and thermal stability were reduced. These findings demonstrate that hydrophobic interactions in Loop1 are essential for maintaining the catalytically competent conformation of TdT, ensuring precise substrate positioning and active site stability. Full article
(This article belongs to the Collection State-of-the-Art Macromolecules in Russia)
Show Figures

Figure 1

15 pages, 3400 KB  
Article
Enhanced Thermostability and Catalytic Efficiency of Alginate Lyase Alyw203 by Hydrogen Bond Network Reconstruction
by Chengcheng Jiang, Jing-Run Ye, Tian-Tian Zhu, Qin Wang, Yan Ma, Zhi-Peng Wang, Chuan-Yang Shi, Ying Wang, Shou-Fu Zhang, Tian-Hong Liu and Hai-Ying Wang
Mar. Drugs 2026, 24(1), 6; https://doi.org/10.3390/md24010006 - 22 Dec 2025
Viewed by 402
Abstract
Alginate lyases are commonly employed for producing alginate oligosaccharides (AOS), but their industrial application is often constrained by low thermal stability and catalytic efficiency. This study engineered mutants of alginate lyase Alyw203 from marine Vibrio based on B-factor values and negative ΔΔG values. [...] Read more.
Alginate lyases are commonly employed for producing alginate oligosaccharides (AOS), but their industrial application is often constrained by low thermal stability and catalytic efficiency. This study engineered mutants of alginate lyase Alyw203 from marine Vibrio based on B-factor values and negative ΔΔG values. The L172V mutant exhibited a 2.43-fold increase in half-life at 40 °C, reduced Km (from 107 to 65 mg/mL), and enhanced kcat/Km (from 0.07 to 0.35 mL/mg/s), indicating improved thermal stability, substrate affinity, and catalytic efficiency. Molecular dynamics simulations revealed that these improvements originated from reconstructed hydrogen bond networks, which stabilized enzyme–substrate interactions and reduced conformational flexibility. These results demonstrate that rational design focused on strengthening hydrogen bonding can simultaneously improve both stability and activity, offering a promising strategy for industrial AOS production. Full article
(This article belongs to the Section Marine Biotechnology Related to Drug Discovery or Production)
Show Figures

Graphical abstract

19 pages, 6410 KB  
Article
β2E153 Residue at Loop B of GABAAR Is Involved in Agonist Stabilization and Gating Properties
by Michał A. Michałowski, Aleksandra Brzóstowicz and Jerzy W. Mozrzymas
Int. J. Mol. Sci. 2026, 27(1), 47; https://doi.org/10.3390/ijms27010047 - 20 Dec 2025
Viewed by 275
Abstract
γ-Aminobutyric acid type A receptors (GABAARs) are pentameric ligand-gated ion channels mediating fast inhibitory neurotransmission in the mammalian brain. Although recent structural and kinetic studies have advanced understandings regarding their activation mechanisms, the molecular determinants coupling agonist binding to channel gating [...] Read more.
γ-Aminobutyric acid type A receptors (GABAARs) are pentameric ligand-gated ion channels mediating fast inhibitory neurotransmission in the mammalian brain. Although recent structural and kinetic studies have advanced understandings regarding their activation mechanisms, the molecular determinants coupling agonist binding to channel gating remain unclear. We investigated the contribution of the β2E153 residue, located on loop B of the extracellular domain, to the activation of α1β2γ2 GABAARs. Macroscopic and single-channel patch clamp recordings were used to characterize two β2E153-mutants: charge reversal (β2E153K) and hydrophobic substitution (β2E153A). Both substitutions disrupted normal receptor kinetics, with β2E153K selectively accelerating deactivation and β2E153A affecting both deactivation and desensitization. Single-channel analysis showed that β2E153A reduced open probability and mean open times, consistent with altered gating transitions inferred from kinetic modeling. Structural inspection suggested that β2E153 forms electrostatic interactions with β2K196 and β2R207 to stabilize loop C and maintain the agonist-bound conformation. The disruption of this interaction likely destabilizes loop C, leading to weakened agonist binding and modified gating. Overall, our results identify β2E153 as a key element in the long-range allosteric network linking the binding site to the channel gate in GABAARs. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

30 pages, 3804 KB  
Article
Evidence Supporting the Hydrophobic-Mismatch Model for Cytochrome b6f-Driven State Transitions in the Cyanobacterium Synechocystis Species PCC 6803
by Terezia Kovacs, Laszlo Kovacs, Mihaly Kis, Michito Tsuyama, Sindhujaa Vajravel, Eva Herman, Nia Petrova, Anelia Dobrikova, Tomas Zakar, Svetla Todinova, Sashka Krumova, Zoltan Gombos and Radka Vladkova
Membranes 2025, 15(12), 383; https://doi.org/10.3390/membranes15120383 - 17 Dec 2025
Viewed by 397
Abstract
While there is a consensus that the cytochrome b6f complex (cytb6f) in algae and plants is involved in the regulatory mechanism of oxygenic photosynthesis known as light-induced state transitions (STs), no such consensus exists for cyanobacteria. Here, [...] Read more.
While there is a consensus that the cytochrome b6f complex (cytb6f) in algae and plants is involved in the regulatory mechanism of oxygenic photosynthesis known as light-induced state transitions (STs), no such consensus exists for cyanobacteria. Here, we provide the first direct functional evidence for cytb6f using single-point mutation data. We introduced a PetD-Phe124Ala substitution in the cyanobacterium Synechocystis sp. PCC 6803 to test the key predictions of the hydrophobic-mismatch (HMM) model for cytb6f-driven STs in all oxygenic photosynthetic species. These predictions concern the role of the Phe/Tyr124fg-loop-PetD and the extent and kinetic characteristics of STs. The effects of PetD-F124A mutation on STs were monitored using 77K and Pulse-Amplitude-Modulated (PAM) fluorescence. For comparison, we employed a phycobilisome (PBS)-less Synechocystis mutant and wild-type (WT) strain, as well as the stn7 mutant and WT of Arabidopsis plant. The PetD-F124A mutation reduced the extent of STs and selectively affected the two-exponential kinetics components of the transitions. Under State 1 conditions, the mutant exhibited ~60% less energetic decoupling of PBS from photosystem I (PSI) compared to the WT. It is explainable by the HMM model with the inability of the PetD-F124A mutant, during the induction phase of the State 2→State 1 transition to adopt the cytb6f conformation with minimal hydrophobic thickness. PAM-derived parameters indicated that PSII electron transport function is not inhibited, and no detectable effect on cyclic electron transport around PSI was observed under low-light conditions. Circular dichroism and differential scanning calorimetry confirmed that both the PSI trimer/monomer ratio and the structural integrity of the PBSs are preserved in the mutant. The compensatory response to the mutation includes decreased PSI content and an increase in PBS rod size. In conclusion, (1) cytb6f is involved in cyanobacterial STs; (2) evidence is provided supporting the HMM model; (3) the electron transfer and signal transduction functions of cytb6f are separated into distinct domains; and (4) the signaling pathway regulating STs and pigment-protein composition in Synechocystis involves PetD-Phe124. Full article
(This article belongs to the Section Biological Membranes)
Show Figures

Figure 1

30 pages, 7533 KB  
Article
Structure-Guided Design of Novel Diarylpyrimidine-Based NNRTIs Through a Comprehensive In Silico Approach: 3D-QSAR, ADMET Evaluation, Molecular Docking, and Molecular Dynamics
by Mouna Baassi, Mohamed Moussaoui, Sanchaita Rajkhowa, Hatim Soufi, Rachid Daoud and Said Belaaouad
Pharmaceuticals 2025, 18(12), 1854; https://doi.org/10.3390/ph18121854 - 5 Dec 2025
Cited by 1 | Viewed by 446
Abstract
Background/Objectives: The emergence of drug-resistant HIV-1 strains challenges the long-term efficacy of current antiretroviral therapies. Non-nucleoside reverse transcriptase inhibitors (NNRTIs) are critical in HIV-1 treatment; however, the need for new candidates with improved resistance profiles and pharmacokinetics remains. This study aims to design [...] Read more.
Background/Objectives: The emergence of drug-resistant HIV-1 strains challenges the long-term efficacy of current antiretroviral therapies. Non-nucleoside reverse transcriptase inhibitors (NNRTIs) are critical in HIV-1 treatment; however, the need for new candidates with improved resistance profiles and pharmacokinetics remains. This study aims to design and evaluate novel NNRTIs targeting both wild-type (WT) and mutant-type (MT) HIV-1 reverse transcriptase (RT) using integrated computational strategies. Methods: We conducted a 3D-QSAR study on 33 naphthyl-diarylpyrimidine derivatives using CoMFA and CoMSIA models. We designed thirty-five novel molecules based on contour map insights. We applied ADMET and drug-likeness filters to prioritize ten candidates. Molecular docking was performed on WT (PDB: 3HVT) and MT (PDB: 4PUO) RT structures. The top candidates underwent 100 ns molecular dynamics (MD) simulations. We analyzed structural stability via RMSD, RMSF, and Rg, while we used SASA and MolSA to assess solvent exposure and surface compactness. Results: The CoMFA and CoMSIA models demonstrated robust predictivity (R2 = 0.979/0.920, Q2 = 0.643/0.546, R2test = 0.747/0.603). P14 and P43 showed higher binding affinities than nevirapine and favorable ADMET profiles. MD simulations confirmed stable binding in WT-RT and adaptive flexibility in MT-RT. SASA and MolSA analysis revealed favorable conformational compaction. Drug-likeness profiles indicated optimal log P, strong hydrogen bonding, and acceptable bioavailability. Conclusions: P14 and P43 demonstrate strong potential as NNRTI leads, combining binding affinity, structural stability, and favorable pharmacokinetics, supporting further experimental development. Full article
(This article belongs to the Special Issue Application of 2D and 3D-QSAR Models in Drug Design)
Show Figures

Graphical abstract

23 pages, 5822 KB  
Article
Computational Analysis of GAT1 Mutations: Functional Consequences from Molecular Dynamics and Binding Free Energy Calculations
by Muhammad Yasir, Jinyoung Park, Eun-Taek Han, Won Sun Park, Jin-Hee Han, Jongseon Choe, Mubashir Hassan, Andrzej Kloczkowski and Wanjoo Chun
Int. J. Mol. Sci. 2025, 26(23), 11339; https://doi.org/10.3390/ijms262311339 - 24 Nov 2025
Cited by 1 | Viewed by 484
Abstract
GABA transporter 1 (GAT1), encoded by the SLC6A1 gene, is essential for maintaining inhibitory neurotransmission by mediating the reuptake of GABA from the synaptic cleft. Dysfunction of GAT1 has been linked to several neurological and neurodevelopmental disorders, including epilepsy and Alzheimer’s disease. In [...] Read more.
GABA transporter 1 (GAT1), encoded by the SLC6A1 gene, is essential for maintaining inhibitory neurotransmission by mediating the reuptake of GABA from the synaptic cleft. Dysfunction of GAT1 has been linked to several neurological and neurodevelopmental disorders, including epilepsy and Alzheimer’s disease. In this study, we performed a comprehensive computational investigation of reported GAT1 mutations to understand their structural and functional implications. Seven mutations (G63S, Y140C, Q291Δ, F294Δ, N310I, D451G, and G457H) were analyzed using homology modeling, structural validation tools, molecular dynamics (MD) simulation triplicates, and binding free energy calculations via the gmx_MMPBSA method. The wild-type consistently exhibited the most favorable interaction energy (−59.89 kcal/mol), the strongest binding free energy (ΔG = −28.29 kcal/mol), and the most stable hydrogen-bonding network. While all mutants displayed elevated RMSD and energy fluctuations relative to the wild-type, these changes predominantly reflected local conformational disturbances rather than global unfolding, indicating that the overall structural framework of GAT1 remains largely preserved. Among the mutants, G63S exerted the mildest effect on ligand stabilization, whereas Y140C, G457H, Q291Δ, and D451G produced substantial reductions in protein–ligand stability, weakened hydrogen bonding, and increased ligand mobility within the binding pocket. Free-energy analysis further highlighted the pronounced destabilizing influence of N310I, Q291Δ, and G457H on tiagabine binding. These findings provide mechanistic insights into how specific GAT1 mutations may alter transporter stability and function, offering a structural framework for future studies on GABAergic dysfunction and therapeutic development. Full article
(This article belongs to the Section Molecular Informatics)
Show Figures

Figure 1

21 pages, 2379 KB  
Article
Deamidation at N53 Causes SOD1 Structural Instability and Excess Zn Incorporation
by Eric Zanderigo, Phyllis Schram, Owen Rogers, Mikayla McLaughlin, Colin A. Smith and Alison L. O’Neil
BioChem 2025, 5(4), 39; https://doi.org/10.3390/biochem5040039 - 13 Nov 2025
Viewed by 553
Abstract
Background/Objectives: Approximately 20% of familial ALS (fALS) cases are linked to mutations in Cu/Zn superoxide dismutase (SOD1). Through a gain function, SOD1 misfolding exerts a toxic effect on motor neurons, leading to their degradation and ALS symptomology in both fALS cases and [...] Read more.
Background/Objectives: Approximately 20% of familial ALS (fALS) cases are linked to mutations in Cu/Zn superoxide dismutase (SOD1). Through a gain function, SOD1 misfolding exerts a toxic effect on motor neurons, leading to their degradation and ALS symptomology in both fALS cases and sporadic ALS (sALS) cases with no known genetic cause. To further our understanding of SOD1-ALS etiology, identifying motor neuron-specific SOD1 post-translational modifications (PTMs) and studying their structural influence is necessary. To this end, we have conducted a study on the influence of the deamidation of Asn53, a PTM proximal to key stabilizing motifs in SOD1, which has scarcely been addressed in the literature to date. Methods: Deamidation to N53 was identified by tandem mass spectrometry of SOD1 immunoprecipitated from motor neuron (MN) cultures derived from wild-type (WT) human induced pluripotent stem cells (iPSCs). WT SOD1 and N53D SOD1, a mutant mimicking the deamidation, were expressed in Escherichia coli and purified for in vitro analyses. Differences between species were measured by experiments probing metal cofactors, relative monomer populations, and aggregation propensity. Furthermore, molecular dynamics experiments were conducted to model and determine the influence of the PTM on SOD1 structure. Results: In contrast to WT, N53D SOD1 showed non-native incorporation of metal cofactors, coordinating more Zn2+ cofactors than total Zn-binding sites, and more readily adopted monomeric forms, unfolded, and aggregated with heating, possibly while releasing coordinated metals. Conclusions: Deamidation to N53 in SOD1 encourages the adoption of non-native conformers, and its detection in WT MN cultures suggests relevance to sALS pathophysiology. Full article
Show Figures

Graphical abstract

26 pages, 1033 KB  
Review
Post-Translational Modifications of Huntingtin: Mechanistic Insights and Therapeutic Opportunities in Huntington’s Disease
by Xiaoxia Zhang, Shengping Zhang and Chuangui Wang
Int. J. Mol. Sci. 2025, 26(22), 10907; https://doi.org/10.3390/ijms262210907 - 11 Nov 2025
Viewed by 1172
Abstract
Huntingtin (HTT) is a large, ubiquitously expressed scaffold protein that participates in multiple cellular processes, including vesicular transport, transcriptional regulation, and energy metabolism. The mutant form of HTT (mHTT), characterized by an abnormal polyglutamine (polyQ) expansion in its N-terminal region, is the causative [...] Read more.
Huntingtin (HTT) is a large, ubiquitously expressed scaffold protein that participates in multiple cellular processes, including vesicular transport, transcriptional regulation, and energy metabolism. The mutant form of HTT (mHTT), characterized by an abnormal polyglutamine (polyQ) expansion in its N-terminal region, is the causative agent of Huntington’s disease (HD), a progressive neurodegenerative disorder. Current therapeutic efforts for HD have primarily focused on lowering HTT levels through gene silencing or promoting mHTT degradation. However, accumulating evidence suggests that post-translational modifications (PTMs) of HTT—such as phosphorylation, ubiquitination, acetylation, and SUMOylation—play pivotal roles in modulating HTT’s conformation, aggregation propensity, subcellular localization, and degradation pathways. These modifications regulate the balance between HTT’s physiological functions and pathological toxicity. Importantly, dysregulation of PTMs has been linked to mHTT accumulation and selective neuronal vulnerability, highlighting their relevance as potential therapeutic targets. A deeper understanding of how individual PTMs and their crosstalk regulate HTT homeostasis may not only provide mechanistic insights into HD pathogenesis but also uncover novel, more specific strategies for intervention. In this review, we summarize recent understanding on HTT PTMs, discuss their implications for disease modification, and outline critical knowledge gaps that remain to be addressed. Full article
(This article belongs to the Special Issue Molecular Insights on Drug Discovery, Design, and Treatment)
Show Figures

Figure 1

15 pages, 7105 KB  
Article
Contribution of the CYP51A Y119F Mutation to Azole Resistance in Aspergillus flavus
by Yabin Zhou, Yue Wang, Alexey A. Grum-Grzhimaylo, Martin Meijer, Bart Kraak, Zhengwen Li and Jos Houbraken
J. Fungi 2025, 11(11), 798; https://doi.org/10.3390/jof11110798 - 10 Nov 2025
Viewed by 865
Abstract
Aspergillus flavus is both an agricultural and clinical pathogen, notable for its ability to contaminate crops with aflatoxins and cause invasive aspergillosis. The increasing emergence of azole resistance in A. flavus poses a serious challenge to food safety and human health. Although mutations [...] Read more.
Aspergillus flavus is both an agricultural and clinical pathogen, notable for its ability to contaminate crops with aflatoxins and cause invasive aspergillosis. The increasing emergence of azole resistance in A. flavus poses a serious challenge to food safety and human health. Although mutations in ergosterol biosynthesis genes have been reported in resistant isolates, their functional contributions remain largely unvalidated. In this study, we investigated the role of the CYP51A Y119F mutation in azole resistance. Site-directed mutants were generated using PCR-based gene editing, and their susceptibility to antifungal agents was assessed through Clinical and Laboratory Standards Institute broth microdilution and agar diffusion assays. The Y119F mutation reduced susceptibility specifically to voriconazole and isavuconazole, while susceptibility to itraconazole and posaconazole remained unchanged. To explore the structural basis of this phenotype, molecular dynamics simulations were performed. The mutant protein exhibited greater fluctuations and reduced conformational stability compared to the wild-type enzyme. Tunnel analysis further indicated that the Y119F substitution caused narrowing and shortening of the main access tunnels to the heme-binding pocket, likely impairing azole access and binding. The combined biochemical and structural analyses suggest that Y119F represents a primary resistance-conferring mutation that modifies the structural dynamics of CYP51A. Full article
(This article belongs to the Section Fungal Pathogenesis and Disease Control)
Show Figures

Figure 1

17 pages, 3747 KB  
Article
Drug Repurposing for AML: Structure-Based Virtual Screening and Molecular Simulations of FDA-Approved Compounds with Polypharmacological Potential
by Mena Abdelsayed and Yassir Boulaamane
Biomedicines 2025, 13(11), 2605; https://doi.org/10.3390/biomedicines13112605 - 24 Oct 2025
Viewed by 1003
Abstract
Background: Acute myeloid leukemia (AML) is a heterogeneous hematologic malignancy characterized by impaired differentiation, apoptosis resistance, and metabolic reprogramming, which collectively contribute to therapeutic resistance and poor clinical outcomes. While targeted agents—such as LSD1 inhibitors, the BCL-2 inhibitor venetoclax, and IDH1 inhibitors—have provided [...] Read more.
Background: Acute myeloid leukemia (AML) is a heterogeneous hematologic malignancy characterized by impaired differentiation, apoptosis resistance, and metabolic reprogramming, which collectively contribute to therapeutic resistance and poor clinical outcomes. While targeted agents—such as LSD1 inhibitors, the BCL-2 inhibitor venetoclax, and IDH1 inhibitors—have provided clinical benefit, their efficacy is often limited by compensatory signaling and clonal evolution. This study aimed to identify FDA-approved compounds with multitarget potential to simultaneously modulate key epigenetic, apoptotic, and metabolic pathways in AML. Methods: Structure-based virtual screening of 3957 FDA-approved molecules was performed against three AML-relevant targets: lysine-specific demethylase 1 (LSD1), BCL-2, and mutant IDH1 (R132H). Top-ranked hits were evaluated using ADMET prediction and molecular dynamics (MD) simulations to assess pharmacokinetic properties, toxicity, and ligand–protein complex stability over 100 ns trajectories. Results: Three compounds—DB16703, DB08512, and DB16047—exhibited high binding affinities across all three targets with favorable pharmacokinetic and safety profiles. MD simulations confirmed the structural stability of the ligand–protein complexes, revealing persistent hydrogen bonding and minimal conformational deviation. These findings suggest that these repurposed drugs possess a promising multitarget profile capable of addressing AML’s multifactorial pathophysiology. Conclusions: This computational study supports the feasibility of a polypharmacology-based strategy for AML therapy by integrating epigenetic modulation, apoptotic reactivation, and metabolic correction within single molecular scaffolds. However, the identified compounds (Belumosudil, DB08512, and Elraglusib) have not yet demonstrated efficacy in AML models; further preclinical validation is warranted to substantiate these predictions and advance translational development. Full article
Show Figures

Figure 1

18 pages, 9504 KB  
Article
Deciphering the Impact of Mutations on PfDHPS Active Site and Sulfadoxine Binding: Structural Insights from Molecular Dynamics Simulations
by Emilie Guémas, Sandie Ménard, Nicolas Jeanne, Georges Landa, Antoine Berry and Marie Brut
Molecules 2025, 30(20), 4118; https://doi.org/10.3390/molecules30204118 - 17 Oct 2025
Viewed by 502
Abstract
The antimalarial combination of sulfadoxine–pyrimethamine is used as a preventive treatment in pregnant women and children in Africa. Sulfadoxine inhibits the Plasmodium falciparum dihydropteroate synthase (PfDHPS), but resistance has emerged through point mutations in this enzyme. In this study, we investigate [...] Read more.
The antimalarial combination of sulfadoxine–pyrimethamine is used as a preventive treatment in pregnant women and children in Africa. Sulfadoxine inhibits the Plasmodium falciparum dihydropteroate synthase (PfDHPS), but resistance has emerged through point mutations in this enzyme. In this study, we investigate the impact of mutations on the structural and dynamic properties of PfDHPS using molecular dynamics simulations. Our results show that PfDHPS maintains overall structural integrity across various combinations of resistance-associated mutations. However, significant differences emerge in ligand binding. Sulfadoxine binding is particularly impacted and shows reduced stability in the mutant systems compared to the wild-type enzyme, while the natural substrate generally maintains stable or even enhanced binding affinity. A key finding is the critical role of the D2 loop, whose conformational flexibility influences ligand retention. In mutant enzymes, the disruption of interactions between the D2 loop and the natural substrate correlates with decreased affinity. In contrast, specific mutations in the loop are associated with an increased affinity. Conversely, sulfadoxine binding is associated with an open D2 loop conformation, facilitating its release from the active site. Finally, the intrinsic flexibility of sulfadoxine emerges as an important determinant of this process. Together, these results provide molecular-level insights into the mechanisms of resistance in PfDHPS and establish a structural and dynamic framework for future investigations into its catalytic function and inhibitor design. Full article
Show Figures

Figure 1

12 pages, 3199 KB  
Article
H128N Substitution in the Sa Antigenic Site of HA1 Causes Antigenic Drift Between Eurasian Avian-like H1N1 and 2009 Pandemic H1N1 Influenza Viruses
by Fei Meng, Zhang Cheng, Zijian Feng, Yijie Zhang, Yali Zhang, Yanwen Wang, Yujia Zhai, Peichun Kuang, Rui Qu, Yan Chen, Chuanling Qiao, Hualan Chen and Huanliang Yang
Viruses 2025, 17(10), 1360; https://doi.org/10.3390/v17101360 - 12 Oct 2025
Viewed by 997
Abstract
The antigenic relationship between Eurasian avian-like H1N1 swine influenza viruses (EA H1N1) and human pandemic 2009 H1N1 viruses (2009/H1N1) remains a critical question for influenza surveillance and vaccine efficacy. This study systematically investigated the antigenic differences between strains A/swine/Tianjin/312/2016 (TJ312, EA H1N1) and [...] Read more.
The antigenic relationship between Eurasian avian-like H1N1 swine influenza viruses (EA H1N1) and human pandemic 2009 H1N1 viruses (2009/H1N1) remains a critical question for influenza surveillance and vaccine efficacy. This study systematically investigated the antigenic differences between strains A/swine/Tianjin/312/2016 (TJ312, EA H1N1) and A/Guangdong-Maonan/SWL1536/2019 (GD1536, 2009/H1N1). Cross-hemagglutination inhibition (HI) assays revealed a significant antigenic disparity, with a 16-fold reduction in heterologous versus homologous HI titers. Comparative sequence analysis identified 22 amino acid differences across the five major antigenic sites (Sa, Sb, Ca1, Ca2, and Cb) of the HA1 subunit. Using reverse genetics, a panel of mutant viruses was generated. This study revealed that a single histidine (H)-to-asparagine (N) substitution at residue 128 (H3 numbering) in the Sa antigenic site acts as a primary determinant of antigenic variation, sufficient to cause a four-fold change in HI titers and a measurable drift in antigenic distance. Structural modeling via AlphaFold3 and PyMOL software suggests that the H128N mutation may alter the local conformation of the antigenic site. It is plausible that H at position 128 could exert electrostatic repulsion with adjacent amino acids, whereas N might facilitate hydrogen bond formation with neighboring residues. These interactions would potentially lead to structural changes in the antigenic site. Our findings confirm that residue 128 is a critical molecular marker for the antigenic differentiation of EA H1N1 and 2009/H1N1 viruses. The study underscores the necessity of monitoring specific HA mutations that could reduce cross-reactivity and provides valuable insights for refining vaccine strain selection and pandemic preparedness strategies. Full article
(This article belongs to the Special Issue Antigenic Drift in Respiratory Viruses)
Show Figures

Figure 1

24 pages, 3369 KB  
Article
The Effects of Heparin Binding and Arg596 Mutations on the Conformation of Thrombin–Antithrombin Michaelis Complex, Revealed by Enhanced Sampling Molecular Dynamics Simulations
by Gábor Balogh and Zsuzsanna Bereczky
Int. J. Mol. Sci. 2025, 26(20), 9901; https://doi.org/10.3390/ijms26209901 - 11 Oct 2025
Viewed by 643
Abstract
The inactivation of thrombin by antithrombin is highly enhanced by the presence of heparin chains forming “bridges” between the two proteins. X-ray structures for such ternary complexes have been published, but the molecular background of the lower efficiency of smaller heparinoids on thrombin [...] Read more.
The inactivation of thrombin by antithrombin is highly enhanced by the presence of heparin chains forming “bridges” between the two proteins. X-ray structures for such ternary complexes have been published, but the molecular background of the lower efficiency of smaller heparinoids on thrombin inhibition remains poorly understood. Antithrombin-resistant prothrombin mutants (mutations affecting Arg596 in prothrombin) have been reported that cause severe thrombophilia. Our aim was to study the interactions in the antithrombin–thrombin Michaelis complex both in the presence and the absence of a heparinoid chain and in the presence of pentasaccharide by using molecular dynamics. We also intended to study the complexes of thrombin mutants as well as a known alternative antithrombin conformation at the “hinge” region built using docking. The binding between the proteins was investigated by Gaussian Accelerated Molecular Dynamics (GaMD). We compared the contribution of several amino acids at the binding “exosites” between AT and the wild type and mutant thrombins and between systems containing or not containing a heparinoid. In the docking-based simulations, several of the analyzed amino acid pairs no longer contributed to the interaction, suggesting that the open “hinge” conformation has limited biological relevance. We could identify multiple conformational types using clustering, revealing high flexibility in mutants and systems without heparinoid, probably indicating lower stability. We were also able to detect the allosteric effects of the ligands on the bound thrombin. In summary, we were able to obtain conformations using GaMD that can explain the better protein–protein interactions in the ternary complexes and the impaired AT binding of the thrombin Arg596 mutants at an atomic level. Full article
(This article belongs to the Special Issue Coagulation Factors and Natural Anticoagulants in Health and Disease)
Show Figures

Figure 1

Back to TopTop