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
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 (596)

Search Parameters:
Keywords = allosteric binding

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 4993 KB  
Article
Stable Non-Competitive DPP-IV Inhibitory Hexapeptide from Parkia timoriana Seeds: A Candidate for Functional Food Development in Type 2 Diabetes
by Sakinah Hilya Abida, Christoper Caesar Yudho Sutopo, Wei-Ting Hung, Nhung Thi Phuong Nong, Tunjung Mahatmanto and Jue-Liang Hsu
Processes 2025, 13(10), 3079; https://doi.org/10.3390/pr13103079 - 26 Sep 2025
Abstract
The tree bean (Parkia timoriana), an underutilized legume valued for its nutritional profile, represents a potential source of bioactive peptides for diabetes management. To our knowledge, this is the first study to identify and characterize DPP-IV inhibitory peptides derived from tree [...] Read more.
The tree bean (Parkia timoriana), an underutilized legume valued for its nutritional profile, represents a potential source of bioactive peptides for diabetes management. To our knowledge, this is the first study to identify and characterize DPP-IV inhibitory peptides derived from tree bean seed protein hydrolysates. The tree bean proteins were digested with trypsin, thermolysin, chymotrypsin, pepsin, and simulated gastrointestinal (SGI) enzymes, among which SGI hydrolysis yielded the highest degree of hydrolysis (14%) and strongest DPP-IV inhibitory activity (IC50 = 1289 ± 58 µg/mL). Guided by DPP-IV inhibitory assays, sequential fractionation using strong cation exchange and RP-HPLC yielded the most potent fraction, H5, with an IC50 of 949 ± 50 µg/mL. After peptide identification and synthesis, APLGPF (AF6) emerged as the most potent inhibitor, with an IC50 of 396 ± 18 µM. Enzyme kinetics revealed a non-competitive inhibition mechanism, corroborated by molecular docking, which indicated binding at an allosteric site of DPP-IV. Furthermore, AF6 remained stable under simulated gastrointestinal digestion and enzymatic exposure, highlighting its resistance to proteolysis. Taken together, these findings highlight P. timoriana as an underexplored source of peptides with DPP-IV inhibitory activity and identify AF6 as a promising lead for developing functional foods or nutraceuticals aimed at type 2 diabetes management. Full article
(This article belongs to the Special Issue Peptides: Advances and Innovations from Discovery to Application)
Show Figures

Graphical abstract

28 pages, 4648 KB  
Article
Allosteric Control Overcomes Steric Limitations for Neutralizing Antibodies Targeting Conserved Binding Epitopes of the SARS-CoV-2 Spike Protein: Exploring the Intersection of Binding, Allostery, and Immune Escape with a Multimodal Computational Approach
by Mohammed Alshahrani, Vedant Parikh, Brandon Foley and Gennady Verkhivker
Biomolecules 2025, 15(9), 1340; https://doi.org/10.3390/biom15091340 - 18 Sep 2025
Viewed by 310
Abstract
Understanding the atomistic basis of multi-layer mechanisms employed by broadly reactive neutralizing antibodies of the SARS-CoV-2 spike protein without directly blocking receptor engagement remains an important challenge in coronavirus immunology. Class 4 antibodies represent an intriguing case: they target a deeply conserved, cryptic [...] Read more.
Understanding the atomistic basis of multi-layer mechanisms employed by broadly reactive neutralizing antibodies of the SARS-CoV-2 spike protein without directly blocking receptor engagement remains an important challenge in coronavirus immunology. Class 4 antibodies represent an intriguing case: they target a deeply conserved, cryptic epitope on the receptor-binding domain yet exhibit variable neutralization potency across subgroups F1 (CR3022, EY6A, COVA1-16), F2 (DH1047), and F3 (S2X259). The molecular basis for this variability is not fully understood. Here, we employed a multi-modal computational approach integrating atomistic and coarse-grained molecular dynamics simulations, binding free energy calculations, mutational scanning, and dynamic network analysis to elucidate how these antibodies engage the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein and influence its function. Our results reveal that neutralization efficacy arises from the interplay of direct interfacial interactions and allosteric effects. Group F1 antibodies (CR3022, EY6A, COVA1-16) primarily operate via classic allostery, modulating flexibility in RBD loop regions to indirectly interfere with the ACE2 receptor binding through long-range effects. Group F2 antibody DH1047 represents an intermediate mechanism, combining partial steric hindrance—through engagement of ACE2-critical residues T376, R408, V503, and Y508—with significant allosteric influence, facilitated by localized communication pathways linking the epitope to the receptor interface. Group F3 antibody S2X259 achieves potent neutralization through a synergistic mechanism involving direct competition with ACE2 and localized allosteric stabilization, albeit with potentially increased escape vulnerability. Dynamic network analysis identified a conserved “allosteric ring” within the RBD core that serves as a structural scaffold for long-range signal propagation, with antibody-specific extensions modulating communication to the ACE2 interface. These findings support a model where Class 4 neutralization strategies evolve through the refinement of peripheral allosteric connections rather than epitope redesign. This study establishes a robust computational framework for understanding the atomistic basis of neutralization activity and immune escape for Class 4 antibodies, highlighting how the interplay of binding energetics, conformational dynamics, and allosteric modulation governs their effectiveness against SARS-CoV-2. Full article
(This article belongs to the Special Issue Protein Biophysics)
Show Figures

Graphical abstract

22 pages, 4007 KB  
Article
Bile Acids Are Potential Negative Allosteric Modulators of M1 Muscarinic Receptors
by Wenbo Yu, Alexander D. MacKerell, David J. Weber and Jean-Pierre Raufman
Biomolecules 2025, 15(9), 1326; https://doi.org/10.3390/biom15091326 - 17 Sep 2025
Viewed by 325
Abstract
The proposed physiological roles of bile acids have expanded beyond the digestion of fats to encompass cell signaling via the activation of a variety of nuclear and plasma membrane receptors in multiple organ systems. The current in silico study was inspired by previous [...] Read more.
The proposed physiological roles of bile acids have expanded beyond the digestion of fats to encompass cell signaling via the activation of a variety of nuclear and plasma membrane receptors in multiple organ systems. The current in silico study was inspired by previous observations from our group and others that bile acids interact functionally with cardiac, pulmonary, and gastrointestinal muscarinic receptors and more recent work demonstrating allosteric binding of cholesterol, the parent molecule for bile acid synthesis, to M1 muscarinic receptors (M1R). Here, we computationally tested the hypothesis that bile acids can allosterically bind to M1R and thereby modulate receptor activation. Utilizing de novo site identification by the ligand competitive saturation (SILCS) method, putative novel allosteric binding sites of bile acid targeting M1R were identified. Molecular dynamics simulations were used to uncover the molecular details of the activation mechanism of M1R due to agonist binding along with allosteric modulation of bile acids on M1R activation. Allosteric binding of bile acids and their glycine and taurine conjugates to M1R negatively impacts the activation process, findings consistent with recent reports that M1R expression and activation inhibit colon cancer cell proliferation. Thus, bile acids may augment colon cancer risk by inhibiting the tumor suppressor actions of M1R. When validated experimentally, these findings are anticipated to shed light on our understanding of how bile acids in the membrane microenvironment can allosterically modulate the function of M1R and possibly other G protein-coupled receptors. Full article
Show Figures

Figure 1

20 pages, 4121 KB  
Article
The Allosteric Communication Network in the Activation of Antithrombin by Heparin
by Gonzalo Izaguirre
Int. J. Mol. Sci. 2025, 26(18), 8984; https://doi.org/10.3390/ijms26188984 - 15 Sep 2025
Viewed by 268
Abstract
The allosteric activation of antithrombin (AT) involves a conformational shift from a native, repressed (R) to a heparin-bound, activated (AH) state. Using computational structural analysis, we identified an evolutionarily conserved allosteric communication network (ACN) comprising the residues H120, Y131, and Y166, which undergo [...] Read more.
The allosteric activation of antithrombin (AT) involves a conformational shift from a native, repressed (R) to a heparin-bound, activated (AH) state. Using computational structural analysis, we identified an evolutionarily conserved allosteric communication network (ACN) comprising the residues H120, Y131, and Y166, which undergo key structural displacements during this transition. Site-directed mutagenesis of these residues markedly enhanced AT native reactivity toward FXa and reduced thermal stability, indicating their role in stabilizing the R state. These findings support a three-step “slingshot” model in which the ACN functions as a molecular lock that restrains stored conformational energy, preventing premature activation. Heparin binding disengages this lock, triggering a cascade of structural changes that propagate from the heparin-binding site (HBS) to the reactive center loop (RCL). Additional mutational analyses of residues bridging the β-sheet A (βsA) and the RCL/exosite domains revealed a delicate energetic balance involving the S380 insertion and E381–R197 salt bridge, which collectively tune the activation threshold. Molecular dynamics simulations of ACN mutants further revealed increased flexibility at both HBS and RCL domains, consistent with concerted allosteric coupling. Together, these results provide new mechanistic insights into the structural basis of AT activation and suggest avenues for engineering heparin-independent AT variants. Full article
(This article belongs to the Special Issue Proteases and Their Inhibitors: From Biochemistry to Applications)
Show Figures

Figure 1

14 pages, 885 KB  
Review
Epigallocatechin Gallate as a Targeted Therapeutic Strategy Against the JAK2V617F Mutation: New Perspectives for the Treatment of Myeloproliferative Neoplasms and Acute Myeloid Leukemia
by Leidivan Sousa Da Cunha, Isabelle Magalhães Farias, Beatriz Maria Dias Nogueira, Caio Bezerra Machado, Flávia Melo Cunha De Pinho Pessoa, Deivide De Sousa Oliveira, Guilherme Passos de Morais, André Pontes Thé, Patrícia Maria Pontes Thé, Manoel Odorico De Moraes Filho, Maria Elisabete Amaral De Moraes and Caroline Aquino Moreira-Nunes
Int. J. Transl. Med. 2025, 5(3), 43; https://doi.org/10.3390/ijtm5030043 - 15 Sep 2025
Viewed by 1870
Abstract
The JAK2V617F mutation is a major molecular factor in Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs) and has been increasingly associated with clonal progression to acute myeloid leukemia (AML), resulting in a poorer prognosis and resistance to conventional therapies. This study integrates a comprehensive literature [...] Read more.
The JAK2V617F mutation is a major molecular factor in Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs) and has been increasingly associated with clonal progression to acute myeloid leukemia (AML), resulting in a poorer prognosis and resistance to conventional therapies. This study integrates a comprehensive literature review with bioinformatic approaches to investigate the potential inhibitory activity of Epigallocatechin Gallate (EGCG), a green tea polyphenol widely recognized for its antioxidant and anticancer properties, on the JAK2V617F mutation. Clinical data from case reports demonstrated heterogeneity in disease progression and frequent therapeutic failures. Molecular docking analysis using the Janus Kinase 2 (JAK2) protein structure (PDB ID: 6D2I) identified a high-affinity binding pocket for EGCG near the V617F mutation site. EGCG exhibited strong binding affinity (−9.2 kcal/mol), forming key interactions with residues Lys581, Ile559, and Leu680, suggesting allosteric modulation of the JH2 pseudokinase domain. To validate our docking protocol, redocking of the known inhibitor AT9283 yielded a favorable Root Mean Square Deviation (RMSD) 2.683 Å and binding energy (−8.3 kcal/mol), confirming the reliability of our approach. Notably, EGCG demonstrated superior binding affinity compared to AT9283 and targets a distinct allosteric site, highlighting its unique mechanism of action and potential as a selective allosteric inhibitor. These findings position EGCG as a promising candidate for future preclinical evaluation, offering a novel strategy to overcome therapy resistance in JAK2V617F-driven malignancies. Full article
Show Figures

Figure 1

25 pages, 1693 KB  
Review
Small-Molecule Ligands of Rhodopsin and Their Therapeutic Potential in Retina Degeneration
by Zaiddodine Pashandi and Beata Jastrzebska
Int. J. Mol. Sci. 2025, 26(18), 8964; https://doi.org/10.3390/ijms26188964 - 15 Sep 2025
Viewed by 414
Abstract
Rhodopsin, the prototypical Class A G protein-coupled receptor (GPCR) and visual pigment of rod photoreceptors, has long served as a structural and mechanistic model for GPCR biology. Mutations in rhodopsin are the leading cause of autosomal dominant retinitis pigmentosa (adRP), making this receptor [...] Read more.
Rhodopsin, the prototypical Class A G protein-coupled receptor (GPCR) and visual pigment of rod photoreceptors, has long served as a structural and mechanistic model for GPCR biology. Mutations in rhodopsin are the leading cause of autosomal dominant retinitis pigmentosa (adRP), making this receptor a critical therapeutic target. In this review, we summarize the chemical, structural, and biophysical features of small-molecule modulators of this receptor, spanning both classical retinoid analogs and emerging non-retinoid scaffolds. These ligands reveal recurrent binding modes within the orthosteric chromophore pocket as well as peripheral allosteric and bitopic sites, where they mediate folding, rescue trafficking, photocycle modulation, and mutant stabilization. We organize ligand performance into a three-tier framework linking binding affinity, cellular rescue potency, and stability gains. Chemotypes in tier 2, which show sub-micromolar to low-micromolar activity with broad mutant coverage, emerge as promising candidates for optimization into next-generation scaffolds. Across scaffolds, a recurring minimal pharmacophore is evident by a contiguous hydrophobic π-surface anchored in the β-ionone region, coupled with a strategically oriented polar handle that modulates the Lys296/Glu113 microenvironment, offering tractable design vectors for non-retinoid chemotypes. Beyond the chromophore binding pocket, we highlight opportunities to exploit extracellular loop epitopes, cytoplasmic microswitch clefts, dimer/membrane interfaces, and ion co-binding sites to engineer safer, state-biased control with fewer photochemical liabilities. By integrating rhodopsin photobiophysics with environment-aware, multi-state medicinal chemistry, and by addressing current translational challenges in drug delivery, this review outlines a rational framework for advancing rhodopsin-targeted therapeutics toward clinically credible interventions for RP and related retinal degenerations. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

16 pages, 1806 KB  
Review
Natural Product-Derived Drugs: Structural Insights into Their Biological Mechanisms
by Yujeong Choi, Younghyun Kim, Hye Joon Boo, Danbi Yoon, Jeong Seok Cha and Jiho Yoo
Biomolecules 2025, 15(9), 1303; https://doi.org/10.3390/biom15091303 - 10 Sep 2025
Viewed by 439
Abstract
Natural product-derived drugs represent a cornerstone of modern pharmacotherapy, with many serving as essential therapeutic agents across diverse medical conditions. Recent advances in structural biology have provided unprecedented insights into the molecular mechanisms underlying their biological activities. This review presents a comprehensive structural [...] Read more.
Natural product-derived drugs represent a cornerstone of modern pharmacotherapy, with many serving as essential therapeutic agents across diverse medical conditions. Recent advances in structural biology have provided unprecedented insights into the molecular mechanisms underlying their biological activities. This review presents a comprehensive structural analysis of five representative natural product-derived drugs: digoxin, simvastatin, morphine, paclitaxel, and penicillin. Through an examination of high-resolution crystal structures and cryo-electron microscopy (cryo-EM) data, we elucidate how these compounds interact with their respective protein targets and modulate biological functions. The structural data reveal diverse binding mechanisms—ranging from competitive inhibition and covalent modification to allosteric modulation via conformational selection and induced fit—demonstrating how natural products achieve their therapeutic effects through precise molecular recognition. These structural insights provide a molecular foundation for understanding natural product pharmacology and offer valuable guidance for structure-based drug design approaches in developing next-generation therapeutics. Full article
Show Figures

Figure 1

16 pages, 6255 KB  
Article
Design of a First-in-Class homoPROTAC to Induce ICP0 Degradation in Human Herpes Simplex Virus 1
by Leyla Salimova, Ali Sahin, Ozge Ardicli, Fatima Hacer Kurtoglu Babayev, Zeynep Betul Sari, Muhammed Emin Sari, Muhammet Guzel Kurtoglu, Sena Ardicli and Huseyn Babayev
Drugs Drug Candidates 2025, 4(3), 42; https://doi.org/10.3390/ddc4030042 - 8 Sep 2025
Viewed by 392
Abstract
Background/Objectives: Human Herpes Simplex Virus 1 (HSV-1) is a common pathogen that establishes lifelong latent infections. The emergence of drug resistance necessitates novel therapeutic strategies. This study introduces a novel antiviral approach: a bivalent degrader designed to induce the degradation of an [...] Read more.
Background/Objectives: Human Herpes Simplex Virus 1 (HSV-1) is a common pathogen that establishes lifelong latent infections. The emergence of drug resistance necessitates novel therapeutic strategies. This study introduces a novel antiviral approach: a bivalent degrader designed to induce the degradation of an essential protein. Methods: A structural model of ICP0, generated via the Chai-1 AI platform, was analyzed with fpocket, P2Rank, and KVFinder to identify a superior allosteric target site. An iterative de novo design workflow with CReM-dock then yielded a lead scaffold based on its predicted affinity and drug-like properties. This selected “warhead” was used to rationally design the final bivalent degrader, ICP0-deg-01, for the ICP0 dimer model. Results: The generative process yielded a lead chemical scaffold that was selected based on its predicted binding affinity and favorable drug-like properties. This scaffold was used to rationally design a single candidate bivalent degrader, ICP0-deg-01. Our structural model predicts that ICP0-deg-01 can successfully bridge two ICP0 protomers, forming an energetically favorable ternary complex. Conclusions: This work provides a computational proof-of-concept for a novel class of anti-herpetic agents and identifies a lead candidate for future molecular dynamics simulations and experimental validation. Full article
(This article belongs to the Section In Silico Approaches in Drug Discovery)
Show Figures

Figure 1

24 pages, 5185 KB  
Article
Lignin-Derived Oligomers as Promising mTOR Inhibitors: Insights from Dynamics Simulations
by Sofia Gabellone, Giovanni Carotenuto, Manuel Arcieri, Paolo Bottoni, Giulia Sbanchi, Tiziana Castrignanò, Davide Piccinino, Chiara Liverani and Raffaele Saladino
Int. J. Mol. Sci. 2025, 26(17), 8728; https://doi.org/10.3390/ijms26178728 - 7 Sep 2025
Viewed by 1521
Abstract
The mammalian target of rapamycin pathway, mTOR, is a crucial signaling pathway that regulates cell growth, proliferation, metabolism, and survival. Due to its dysregulation it is involved in several ailments such as cancer or age-related diseases. The discovery of mTOR and the understanding [...] Read more.
The mammalian target of rapamycin pathway, mTOR, is a crucial signaling pathway that regulates cell growth, proliferation, metabolism, and survival. Due to its dysregulation it is involved in several ailments such as cancer or age-related diseases. The discovery of mTOR and the understanding of its biological functions were greatly facilitated by the use of rapamycin, an antibiotic of natural origin, which allosterically inhibits mTORC1, effectively blocking its function. In this entirely computational study, we investigated mTOR’s interaction with seven ligands: two clinically established inhibitors (everolimus and rapamycin) and five lignin-derived oligomers, a renewable natural polyphenol recently used for the drug delivery of everolimus. The seven complexes were analyzed through all-atom molecular dynamics simulations in explicit solvent using a high-performance computing platform. Trajectory analyses revealed stable interactions between mTOR and all ligands, with lignin-derived compounds showing comparable or enhanced binding stability relative to reference drugs. To evaluate the stability of the molecular complex and the behavior of the ligand over time, we analyzed key parameters including root mean square deviation, root mean square fluctuation, number of hydrogen bonds, binding free energy, and conformational dynamics assessed through principal component analysis. Our results suggest that lignin fragments are a promising, sustainable scaffold for developing novel mTOR inhibitors. Full article
(This article belongs to the Special Issue The Application of Machine Learning to Molecular Dynamics Simulations)
Show Figures

Figure 1

14 pages, 2877 KB  
Article
Ivermectin Binds to the Allosteric Site (Site 2) and Inhibits Allosteric Integrin Activation by TNF and Other Pro-Inflammatory Cytokines
by Yoko K. Takada and Yoshikazu Takada
Int. J. Mol. Sci. 2025, 26(17), 8655; https://doi.org/10.3390/ijms26178655 - 5 Sep 2025
Viewed by 820
Abstract
Ivermectin (IVM), a broad-spectrum anthelmintic agent, has anti-inflammatory properties, and affects cellular and humoral immune responses. We recently showed that multiple pro-inflammatory cytokines (e.g., FGF2, CCL5, CD40L) bind to the allosteric site (site 2) of integrins and activate them. 25-Hydroxycholesterol, a pro-inflammatory lipid [...] Read more.
Ivermectin (IVM), a broad-spectrum anthelmintic agent, has anti-inflammatory properties, and affects cellular and humoral immune responses. We recently showed that multiple pro-inflammatory cytokines (e.g., FGF2, CCL5, CD40L) bind to the allosteric site (site 2) of integrins and activate them. 25-Hydroxycholesterol, a pro-inflammatory lipid mediator, is known to bind to site 2 and induce integrin activation and inflammatory signals (e.g., IL-6 and TNF secretion), suggesting that site 2 is critically involved in inflammation. We showed that two anti-inflammatory cytokines (FGF1 and NRG1) bind to site 2 and inhibit integrin activation by inflammatory cytokines. We hypothesized that ivermectin binds to site 2 and inhibits inflammatory signaling by pro-inflammatory cytokines. A docking simulation predicts that ivermectin binds to site 2. Ivermectin inhibits the integrin activation induced by inflammatory cytokines, suggesting that ivermectin is a site 2 antagonist. We showed that TNF, a major pro-inflammatory cytokine, binds to integrin site 2 and induces allosteric integrin activation like other pro-inflammatory cytokines, suggesting that site 2 binding and integrin activation is a potential mechanism of the pro-inflammatory action of these cytokines. Ivermectin suppressed the activation of soluble β3 integrins by TNF and other pro-inflammatory cytokines in a dose-dependent manner in cell-free conditions. Binding to site 2 and the inhibition of binding of inflammatory cytokines may be a potential mechanism of anti-inflammatory action of ivermectin. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

16 pages, 2565 KB  
Article
Pharmacological Characterization of the Novel CRF1 Receptor Antagonist, Thiazolo[4,5-d] Pyrimidine Analog, M43
by Spyridon Marios Giatro, George Komontachakis, Aikaterini Kalantidou, Nastazia Lesgidou, Vlasios Karageorgos, Mohamed Teleb, Md Rabiul Islam, Thomas Mavromoustakos, Hesham Fahmy, Maria Venihaki, Minos-Timotheos Matsoukas and George Liapakis
Biomolecules 2025, 15(9), 1265; https://doi.org/10.3390/biom15091265 - 1 Sep 2025
Viewed by 664
Abstract
The corticotropin-releasing factor (CRF) and its type 1 receptor (CRF1R) play a key role in the regulation of the hypothalamic–pituitary–adrenal (HPA) axis. Dysregulation of the HPA axis is associated with congenital adrenal hyperplasia (CAH) and depression. Non-peptide CRF1R-selective antagonists [...] Read more.
The corticotropin-releasing factor (CRF) and its type 1 receptor (CRF1R) play a key role in the regulation of the hypothalamic–pituitary–adrenal (HPA) axis. Dysregulation of the HPA axis is associated with congenital adrenal hyperplasia (CAH) and depression. Non-peptide CRF1R-selective antagonists displayed antidepressant effects on animal models and are used for the management of CAH. To develop novel non-peptide CRF1R antagonists, we have previously designed and synthesized a series of substituted pyrimidines. Among these analogs, molecule 43 (M43) binds to CRF1R with the highest affinity. Based on this finding, we selected M43 for further pharmacological characterization in the present study. The results suggest that M43 is a potent CRF1R antagonist, blocking the ability of the CRF-related agonist, Tyr0-sauvagine, to stimulate (1) cAMP accumulation in HEK 293 cells expressing CRF1R and (2) the proliferation rate of RAW 264.7 macrophages. Computational studies suggest that the antagonist properties of M43 are mostly attributed to its ability to interact with residues in the allosteric pocket of CRF1R, comprised of the third, fifth, and sixth transmembrane domain residues, which block activation-associated structural rearrangements of the receptor. Our data will be used to design novel non-peptide CRF1R antagonists for clinical use. Full article
Show Figures

Graphical abstract

28 pages, 5850 KB  
Article
Integrated Approach for Biochemical and Functional Characterization of Six Clinical Variants of Glucose-6-Phosphate Dehydrogenase
by Beatriz Hernández-Ochoa, Mónica Guadalupe Gualos-González, Jhuremy Alexandra Moreno-Hernández, Laura Morales-Luna, Montserrat Vázquez-Bautista, Luis Miguel Canseco-Ávila, Verónica Pérez de la Cruz, Roberto Arreguin-Espinosa, Elizabeth Hernández-Urzua, Sergio Enríquez-Flores, Ignacio De la Mora-De la Mora, Noemí Cárdenas-Rodríguez, Cindy Bandala, Lucia De Franceschi, Abraham Vidal-Limon and Saúl Gómez-Manzo
Int. J. Mol. Sci. 2025, 26(17), 8464; https://doi.org/10.3390/ijms26178464 - 30 Aug 2025
Viewed by 518
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) deficiency is a widespread enzymopathy affecting approximately 500 million individuals that represents a significant global health issue. Among the more than 230 identified mutations in the G6PD gene, six class A variants—G6PD Utrecht (Pro409Ser), G6PD Suwalki (Pro409Arg), G6PD Merlo (Pro409Gln), [...] Read more.
Glucose-6-phosphate dehydrogenase (G6PD) deficiency is a widespread enzymopathy affecting approximately 500 million individuals that represents a significant global health issue. Among the more than 230 identified mutations in the G6PD gene, six class A variants—G6PD Utrecht (Pro409Ser), G6PD Suwalki (Pro409Arg), G6PD Merlo (Pro409Gln), G6PD Kawasaki (Gly410Ala), G6PD Shinagawa (Gly410Asp), and G6PD Riverside (Gly410Cys)—are located in the beta-loop near the NADP+ binding site. These mutations are of particular interest due to their association with severe hematologic phenotypes, including chronic hemolytic anemia, as well as their proposed role in the allosteric regulation of G6PD multimerization. This study presents a comprehensive biochemical and functional characterization of these clinically relevant G6PD variants. The variant enzymes were cloned, expressed, and purified for characterization. Kinetic parameters and thermal stability assays, complemented by molecular dynamics simulations (MDS), were employed to elucidate the structural impacts of the mutations. Our results demonstrate that these mutations significantly impair protein function, characterized by reduced affinity for glucose-6-phosphate (G6P) and NADP+, as well as altered thermal stability compared with wild-type G6PD. MDS revealed that point mutations in the βN- and βM-sheets in the NADP+s region propagate subtle conformational changes, ultimately affecting the NADP+c region and the G6P binding cavity. Furthermore, secondary structure element analyses of the simulation data showed that Pro409 and Gly410 point mutations propagate several changes around residues 195–210 (G6P binding site) and 380–400 (NADP+s), explaining their effect on overall catalytic performance. These findings enhance our understanding of the molecular mechanisms underlying G6PD deficiency and its clinical implications, providing a foundation for future therapeutic strategies aimed at mitigating the effects of these variants. Full article
(This article belongs to the Special Issue The Application of Machine Learning to Molecular Dynamics Simulations)
Show Figures

Figure 1

24 pages, 15675 KB  
Article
Meaty Aroma Compounds Enhance MSG Umami Perception Through Allosteric Modulation of T1R1/T1R3 Receptor: Evidence from Nasal Clip Sensory Evaluation and Molecular Dynamics Simulation
by Yaqi Zhao, Jianan Zhang, Mouming Zhao, Xuan Zhao and Guowan Su
Foods 2025, 14(17), 3041; https://doi.org/10.3390/foods14173041 - 29 Aug 2025
Viewed by 644
Abstract
Background: Understanding how aroma compounds enhance monosodium glutamate (MSG) umami perception remains a critical challenge in flavor science. Methods: The umami-enhancing effects of meaty flavorings were investigated using nasal clip sensory evaluation (orthonasal blockage). Active aroma compounds were subsequently identified using gas chromatography-mass [...] Read more.
Background: Understanding how aroma compounds enhance monosodium glutamate (MSG) umami perception remains a critical challenge in flavor science. Methods: The umami-enhancing effects of meaty flavorings were investigated using nasal clip sensory evaluation (orthonasal blockage). Active aroma compounds were subsequently identified using gas chromatography-mass spectrometry (GC-MS). The three-dimensional structure of the umami receptor T1R1/T1R3 was constructed by homology modeling. The interaction mechanism was deciphered using molecular dynamics (MD) simulations. Results: Seafood essence S demonstrated the most potent umami enhancement. Five key compounds significantly intensified the MSG umami intensity: methional, dimethyl sulfide (DMS), D-limonene (DLE), 2,3-dimethylpyrazine, and dimethyl trisulfide. Notably, this enhancement persisted even under nasal clip conditions, revealing a novel mechanism independent of cross-modal interactions. Sulfur-containing compounds consistently demonstrated umami-enhancing effects across the evaluation conditions. MD simulations showed that aroma compounds induced allosteric remodeling of T1R1/T1R3, strengthening MSG-receptor hydrogen bonding (1.8–2.6-fold increase), reducing receptor flexibility, and stabilizing the ternary complex. Binding affinity was highest for DMS, followed by DLE and methional. Conclusion: This study provides the first receptor-level evidence that aroma compounds directly modulate MSG-taste receptor interactions through allosteric regulation, offering a novel theoretical framework for odor–taste interactions with significant implications for umami enhancer design and flavor research. Full article
Show Figures

Graphical abstract

19 pages, 3937 KB  
Article
Exploring the Allosteric Pathways of Asciminib in the Dual Inhibition of BCR-ABL1
by Jie Ming, Hongwei Gao and Jiuyu Zhan
Biomolecules 2025, 15(9), 1214; https://doi.org/10.3390/biom15091214 - 22 Aug 2025
Viewed by 698
Abstract
The BCR-ABL1 fusion protein is a critical therapeutic target in Chronic Myeloid Leukemia (CML). Current monotherapy approaches involve types of inhibitors that can be categorized into ATP competitive inhibitors and allosteric inhibitors. However, resistance mutations in the tyrosine kinase domain of BCR-ABL1 have [...] Read more.
The BCR-ABL1 fusion protein is a critical therapeutic target in Chronic Myeloid Leukemia (CML). Current monotherapy approaches involve types of inhibitors that can be categorized into ATP competitive inhibitors and allosteric inhibitors. However, resistance mutations in the tyrosine kinase domain of BCR-ABL1 have limited the effectiveness of these drugs. Research indicates that dual inhibition of BCR-ABL1 by combining these two types of inhibitors effectively addresses the issue of drug resistance as there are no overlapping resistance mechanisms. However, the underlying reasons for the observed synergistic effects have not yet been thoroughly elucidated. In this study, we employed molecular dynamics simulation to observe the synergistic interactions of BCR-ABL1 by the allosteric inhibitor asciminib and ATP competitive inhibitors nilotinib and ponatinib. Our study reveals that when asciminib binds to BCR-ABL1, nilotinib and ponatinib exhibit more substantial binding stability compared to monotherapy. At the atomic level, we have elucidated the reasons for the enhanced binding affinity of nilotinib and ponatinib when using a co-inhibition therapy. Our study reveals the allosteric communication pathway between asciminib and ponatinib, providing more detailed insights into the effectiveness of combination therapy. These findings provide valuable insights into combination therapies, aiding in the rational use of medications and guiding the design of novel inhibitors. Full article
Show Figures

Figure 1

13 pages, 1653 KB  
Article
Dose-Dependent Dual Effect of the Endozepine ODN on Neuronal Spiking Activity
by Mahmoud Hazime, Marion Gasselin, Michael Alasoadura, Juliette Leclerc, Benjamin Lefranc, Magali Basille-Dugay, Celine Duparc, David Vaudry, Jérôme Leprince and Julien Chuquet
Brain Sci. 2025, 15(8), 885; https://doi.org/10.3390/brainsci15080885 - 20 Aug 2025
Viewed by 596
Abstract
Background/Objectives: Endozepines known as the endogenous ligands of benzodiazepine-binding sites, include the diazepam binding inhibitor (DBI) and its processing products, the triakontatetraneuropeptide (TTN) and the octadecaneuropeptide (ODN). Despite indisputable evidence of the binding of ODN on GABAAR-BZ-binding sites, their action on [...] Read more.
Background/Objectives: Endozepines known as the endogenous ligands of benzodiazepine-binding sites, include the diazepam binding inhibitor (DBI) and its processing products, the triakontatetraneuropeptide (TTN) and the octadecaneuropeptide (ODN). Despite indisputable evidence of the binding of ODN on GABAAR-BZ-binding sites, their action on this receptor lacks compelling electrophysiological observations, with some studies reporting that ODN acts as a negative allosteric modulator (NAM) of GABAAR while others suggest the opposite (positive allosteric modulation, PAM effect). All these studies were carried out in vitro with various neuronal cell types. To further elucidate the role of ODN in neuronal excitability, we tested its effect in vivo in the cerebral cortex of the anesthetized mouse. Methods: Spontaneous neuronal spikes were recorded by means of an extracellular pipette, in the vicinity of which ODN was micro-infused, either at a high dose (10−5 M) or low dose (10−11 M). Results: ODN at a high dose induced a significant increase in neuronal spiking. This effect could be antagonized by the GABAAR-BZ-binding site blocker flumazenil. In sharp contrast, at low concentrations, ODN reduced neuronal spiking with a magnitude similar to GABA itself. Interestingly, this decrease in neuronal activity by low dose of ODN was not flumazenil-dependent, suggesting that this effect is mediated by another receptor. Finally, we show that astrocytes in culture, known to be stimulated by picomolar doses of ODN via a GPCR, increased their export of GABA when stimulated by low dose of ODN. Conclusion: Our results confirm the versatility of ODN in the control of GABA transmission, but suggest that its PAM-like effect is, at least in part, mediated via an astrocytic non-GABAAR ODN receptor release of GABA. Full article
(This article belongs to the Section Neuroglia)
Show Figures

Figure 1

Back to TopTop