151
|
Anabtawi N, Nicolet D, Alotaibi N, Buelow DR, Orwick S, Gregory T, Raj R, Coleman K, Kolitz JE, Powell BL, Blum WG, Baer MR, Byrd JC, Stock W, Uy GL, Mrózek K, Eisfeld AK, Cheng X, Baker SD, Blachly JS. Prognostic, biological, and structural implications of FLT3-JMD point mutations in acute myeloid leukemia: an analysis of Alliance studies. Leukemia 2025; 39:623-631. [PMID: 39806020 PMCID: PMC11879849 DOI: 10.1038/s41375-024-02498-y] [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: 11/20/2024] [Revised: 11/22/2024] [Accepted: 12/04/2024] [Indexed: 01/16/2025]
Abstract
The FLT3 gene frequently undergoes mutations in acute myeloid leukemia (AML), with internal tandem duplications (ITD) and tyrosine kinase domain (TKD) point mutations (PMs) being most common. Recently, PMs and deletions in the FLT3 juxtamembrane domain (JMD) have been identified, but their biological and clinical significance remains poorly understood. We analyzed 1660 patients with de novo AML and found FLT3-JMD mutations, mostly PMs, in 2% of the patients. Patients with FLT3-JMD mutations had a higher relapse rate and shorter disease-free survival than those with FLT3-TKD, whereas their relapse rate, disease-free and overall survival were not significantly different from those of FLT3-ITD-positive patients. In vitro experiments showed that FLT3-JMD PMs transformed hematopoietic cells and responded well to type I and II FLT3 inhibitors. Molecular dynamics simulations were used to explore the conformational changes of JMD PMs relative to wild-type FLT3. These mutations exhibited constrained domain motions with wider gate openings, potentially enhancing drug binding. Altered residue interactions and structural changes shed light on their unique functional mechanisms, with increased allosteric pathways suggesting reduced interactions with other residues. We conclude that patients with FLT3-JMD PMs represent uncommon but important subset with distinct molecular and biological features, and may benefit from FLT3 inhibitors.
Collapse
Affiliation(s)
- Nadeen Anabtawi
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Deedra Nicolet
- The Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- Alliance Statistics and Data Management Center, The Ohio State University, Columbus, OH, USA
| | - Najla Alotaibi
- The Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Daelynn R Buelow
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Shelley Orwick
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Thomas Gregory
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Ruchika Raj
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Kennedy Coleman
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Jonathan E Kolitz
- Monter Cancer Center, Zucker School of Medicine at Hofstra/Northwell, Lake Success, NY, USA
- Monter Cancer Center, North Shore University Hospital and Long Island Jewish Medical Center, Northwell Health, New Hyde Park, NY, USA
| | - Bayard L Powell
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston Salem, NC, USA
| | - William G Blum
- Department of Hematology and Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Maria R Baer
- University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - John C Byrd
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Wendy Stock
- Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Geoffrey L Uy
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Krzysztof Mrózek
- The Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Ann-Kathrin Eisfeld
- The Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Xiaolin Cheng
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Sharyn D Baker
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA.
| | - James S Blachly
- The Clara D. Bloomfield Center for Leukemia Outcomes Research, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
152
|
Rasul HO, Ghafour DD, Aziz BK, Hassan BA, Rashid TA, Kivrak A. Decoding Drug Discovery: Exploring A-to-Z In Silico Methods for Beginners. Appl Biochem Biotechnol 2025; 197:1453-1503. [PMID: 39630336 DOI: 10.1007/s12010-024-05110-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2024] [Indexed: 03/29/2025]
Abstract
The drug development process is a critical challenge in the pharmaceutical industry due to its time-consuming nature and the need to discover new drug potentials to address various ailments. The initial step in drug development, drug target identification, often consumes considerable time. While valid, traditional methods such as in vivo and in vitro approaches are limited in their ability to analyze vast amounts of data efficiently, leading to wasteful outcomes. To expedite and streamline drug development, an increasing reliance on computer-aided drug design (CADD) approaches has merged. These sophisticated in silico methods offer a promising avenue for efficiently identifying viable drug candidates, thus providing pharmaceutical firms with significant opportunities to uncover new prospective drug targets. The main goal of this work is to review in silico methods used in the drug development process with a focus on identifying therapeutic targets linked to specific diseases at the genetic or protein level. This article thoroughly discusses A-to-Z in silico techniques, which are essential for identifying the targets of bioactive compounds and their potential therapeutic effects. This review intends to improve drug discovery processes by illuminating the state of these cutting-edge approaches, thereby maximizing the effectiveness and duration of clinical trials for novel drug target investigation.
Collapse
Affiliation(s)
- Hezha O Rasul
- Department of Pharmaceutical Chemistry, College of Science, Charmo University, Peshawa Street, Chamchamal, 46023, Sulaimani, Iraq.
| | - Dlzar D Ghafour
- Department of Medical Laboratory Science, College of Science, Komar University of Science and Technology, 46001, Sulaimani, Iraq
- Department of Chemistry, College of Science, University of Sulaimani, 46001, Sulaimani, Iraq
| | - Bakhtyar K Aziz
- Department of Nanoscience and Applied Chemistry, College of Science, Charmo University, Peshawa Street, Chamchamal, 46023, Sulaimani, Iraq
| | - Bryar A Hassan
- Computer Science and Engineering Department, School of Science and Engineering, University of Kurdistan Hewler, KRI, Iraq
- Department of Computer Science, College of Science, Charmo University, Peshawa Street, Chamchamal, 46023, Sulaimani, Iraq
| | - Tarik A Rashid
- Computer Science and Engineering Department, School of Science and Engineering, University of Kurdistan Hewler, KRI, Iraq
| | - Arif Kivrak
- Department of Chemistry, Faculty of Sciences and Arts, Eskisehir Osmangazi University, Eskişehir, 26040, Turkey
| |
Collapse
|
153
|
El Hage K, Dhayalan B, Chen Y, Phillips NB, Whittaker J, Carr K, Whittaker L, Phillips MH, Ismail‐Beigi F, Meuwly M, Weiss MA. Stabilization of a protein by a single halogen-based aromatic amplifier. Protein Sci 2025; 34:e70064. [PMID: 39969055 PMCID: PMC11837044 DOI: 10.1002/pro.70064] [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: 08/23/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/20/2025]
Abstract
The utility of halogenation in protein design is investigated by a combination of quantitative atomistic simulations and experiment. Application to insulin is of complementary basic and translational interest. In a singly halogenated aromatic ring, regiospecific inductive effects were predicted to modulate multiple surrounding electrostatic (weakly polar) interactions, thereby amplifying changes in thermodynamic stability. In accordance with the simulations, we demonstrated stabilization of insulin by single halogen atoms at the ortho position of an invariant phenylalanine (2-F-PheB24, 2-Cl-PheB24, and 2-Br-PheB24; ΔΔGu = -0.5 to -1.0 kcal/mol) located at the edge of a protein crevice; corresponding meta and para substitutions had negligible effects. Although receptor-binding affinities were generally decreased (in accordance with packing of the native Phe at the hormone-receptor interface), the ortho-analogs retained biological activity in mammalian cells and in a rat model of diabetes mellitus. Further, the ortho-modified analogs exhibited enhanced resistance to fibrillation above room temperature in two distinct assays of physical stability. Regiospecific halo-aromatic stabilization may thus augment the shelf life of pharmaceutical insulin formulations under real-world conditions. This approach, extending principles of medicinal chemistry, promises to apply to a broad range of therapeutic proteins and vaccines whose biophysical stabilization would enhance accessibility in the developing world.
Collapse
Affiliation(s)
- Krystel El Hage
- Department of ChemistryUniversity of BaselBaselSwitzerland
- Qubit PharmaceuticalsParisFrance
| | - Balamurugan Dhayalan
- Department of Biochemistry & Molecular BiologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Yen‐Shan Chen
- Department of Biochemistry & Molecular BiologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Nelson B. Phillips
- Department of BiochemistryCase Western Reserve UniversityClevelandOhioUSA
| | - Jonathan Whittaker
- Department of BiochemistryCase Western Reserve UniversityClevelandOhioUSA
| | - Kelley Carr
- Department of BiochemistryCase Western Reserve UniversityClevelandOhioUSA
| | - Linda Whittaker
- Department of BiochemistryCase Western Reserve UniversityClevelandOhioUSA
| | | | - Faramarz Ismail‐Beigi
- Department of BiochemistryCase Western Reserve UniversityClevelandOhioUSA
- Department of MedicineCase Western Reserve UniversityClevelandOhioUSA
| | - Markus Meuwly
- Department of ChemistryUniversity of BaselBaselSwitzerland
| | - Michael A. Weiss
- Department of Biochemistry & Molecular BiologyIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
154
|
Kumar P, Kumar V, Sharma S, Sharma R, Warghat AR. Fritillaria steroidal alkaloids and their multi-target therapeutic mechanisms: insights from network pharmacology. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2209-2228. [PMID: 39382678 DOI: 10.1007/s00210-024-03502-z] [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: 08/20/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024]
Abstract
Medicinal Fritillaria herbs, known for their rich content of steroidal alkaloids, have emerged as promising candidates in the treatment of chronic diseases due to their diverse pharmacological properties. Leveraging advancements in network pharmacology and molecular docking, this study explores the multi-target mechanisms through which these alkaloids exert therapeutic effects. The integration of bioinformatics, systems biology, and pharmacology in drug discovery has provided insights into the molecular interactions and pathways influenced by Fritillaria steroidal alkaloids. This review synthesizes comprehensive literature from 1985 to 2024, revealing the potential of these compounds in addressing respiratory diseases, inflammation, and cancer. The integration of traditional Chinese medicine (TCM) with modern pharmacological techniques underscores the relevance of these compounds in next-generation drug discovery. While initial findings are promising, further empirical validation is necessary to fully harness the therapeutic potential of Fritillaria steroidal alkaloids.
Collapse
Affiliation(s)
- Pankaj Kumar
- Department of Biotechnology, Dr Y.S, Parmar University of Horticulture and Forestry, Solan, Himachal Pradesh, India.
| | - Vinay Kumar
- Department of Biotechnology, Dr Y.S, Parmar University of Horticulture and Forestry, Solan, Himachal Pradesh, India
| | - Shagun Sharma
- Department of Biotechnology, Dr Y.S, Parmar University of Horticulture and Forestry, Solan, Himachal Pradesh, India
| | - Rohit Sharma
- Department of Forest Products, Dr Y.S, Parmar University of Horticulture and Forestry, Solan, Himachal Pradesh, India
| | - Ashish R Warghat
- CSIR-Institute of Himalayan Bioresource Technology, Palampur, 176061, Himachal Pradesh, India
- Academy of Scientific and Innovative Research, Ghaziabad, 201002, India
| |
Collapse
|
155
|
Ogunlakin AD, Ojo OA, Gyebi GA, Akinwumi IA, Adebodun GO, Ayokunle DI, Ambali OA, Ayeni PO, Awosola OE, Babatunde DE, Akintunde EA, Ajayi-Odoko OA, Dahunsi OS, Sonibare MA. Elemental evaluation, nutritional analysis, GC-MS analysis and ameliorative effects of Artocarpus communis J.R.Forst. & G.Forst. seeds' phytoconstituents on metabolic syndrome via in silico approach. J Biomol Struct Dyn 2025; 43:1981-2001. [PMID: 38112300 DOI: 10.1080/07391102.2023.2293271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 12/01/2023] [Indexed: 12/21/2023]
Abstract
The nutritional as well as beneficial effects of the Artocarpus communis seed on metabolic syndrome complications have not been studied. In this research, the aim was to investigate the nutritional composition and beneficial effects of Artocarpus communis seeds' phytoconstituents on the p53 core, fat mass and obesity-associated (FTO) protein and cytochrome P450 CYP11A1 domains. The elements and phytochemicals in the seed were determined through atomic absorption spectroscopy assay and gas chromatography-mass spectrometry (GC-MS) analysis, respectively. Also, the compounds detected were docked to the p53 core, FTO protein and cytochrome P450 CYP11A1 domains protein. Artocarpus communis seed contains sodium (7.824 ± 0.0134 ppm), magnesium (10.187 ± 0.0239 ppm) and iron (1.924 ± 0.0017), while zinc and cadmium were undetected. Phenolics and flavonoids were the most abundant phytochemicals in the seed. Phytoconstituents, such as pentadecanoic acid, hexadecanoic acid and methyl ester, possessing different therapeutic effects were identified via GC-MS analysis. In A. communis seed, 3-methyl-4-nitro-5-(1-pyrazolyl) pyrazole and phenanthrene were able to bind more peculiarly and specifically to the p53 core, FTO protein and cytochrome P450 CYP11A1 domains. One of the important processes that were hypothesized for the recovery of metabolic syndrome in affected victims is shown by the molecular dynamics analysis, which shows that the binding of these chemicals to the targeted structure stabilized the proteins. Therefore, Artocarpus communis seeds could be a new strategy for the management of metabolic syndrome.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Akingbolabo Daniel Ogunlakin
- Bowen University SDG 03 (Good Health and Wellbeing Research Cluster), Iwo, Nigeria
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Laboratory (PMTCB-RL), Department of Biochemistry, Bowen University, Iwo, Nigeria
| | - Oluwafemi Adeleke Ojo
- Bowen University SDG 03 (Good Health and Wellbeing Research Cluster), Iwo, Nigeria
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Laboratory (PMTCB-RL), Department of Biochemistry, Bowen University, Iwo, Nigeria
| | - Gideon Ampoma Gyebi
- Natural Products and Structural (Bio-Chem)-Informatics Research Laboratory (NpsBC-RI), Department of Biochemistry, Bingham University, Karu, Nigeria
| | | | | | | | - Owoola Azeezat Ambali
- School of Medicine and Allied Health Sciences, University of the Gambia, Banjul, Gambia
| | - Peluola Olujide Ayeni
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Laboratory (PMTCB-RL), Department of Biochemistry, Bowen University, Iwo, Nigeria
| | | | | | | | | | | | - Mubo Adeola Sonibare
- Department of Pharmacognosy, Faculty of Pharmacy, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
156
|
Magar HS, Fayez M, Febbraio F, Hassan RYA. Esterase-2 mutant-based nanostructured amperometric biosensors for the selective determination of paraoxon (Neurotoxin). Anal Biochem 2025; 698:115751. [PMID: 39681172 DOI: 10.1016/j.ab.2024.115751] [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: 10/08/2024] [Revised: 11/29/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024]
Abstract
Organophosphate pesticides (OPs) are causing non-selective inhibition in enzymatic bioreceptors, thus the enzymatic-inhibition-based traditional assays are not suitable for their specific detection in food and environmental samples. Accordingly, a selective nanostructured electrochemical biosensing system was designed using six mutants of the esterase-2 (EST2 protein) enzymes from A. acidocaldarius to be exploited as targeting bio-receptors for the specific detection of OPs. Each of the EST2 mutant enzymes was immobilized on disposable screen-printed electrodes modified with Aluminum oxide (Al2O3)/Copper (Cu) nanocomposite. Consequently, chronoamperometric assay was fully optimized, and cross-reactivity study was carried out using paraoxon, malathion and chlorpyrifos. The comparative cross-reactivity study was performed on the six mutant proteins in terms of inhibitory percentage over a wide range of pesticide concentrations. Eventually, a wide dynamic inhibition range was achieved while the limit of detection for the paraoxon toxicity was 0.01 nM and the limit of quantification was 0.05 nM. Finally, paraoxon was selectively determined using the newly developed EST-based biosensor in different spiked food samples.
Collapse
Affiliation(s)
- Hend Samy Magar
- Applied Organic Chemistry Department, National Research Centre (NRC), Dokki, Giza, 2622, Egypt
| | - Muhammad Fayez
- Biosensors Research Lab, Zewail City of Science and Technology, 6th October City, Giza, 12578, Egypt
| | - Ferdinando Febbraio
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), Via P. Castellino 111, 80131, Naples, Italy.
| | - Rabeay Y A Hassan
- Biosensors Research Lab, Zewail City of Science and Technology, 6th October City, Giza, 12578, Egypt; Applied Organic Chemistry Department, National Research Centre (NRC), Dokki, Giza, 2622, Egypt.
| |
Collapse
|
157
|
Le MHN, Nguyen PK, Nguyen TPT, Nguyen HQ, Tam DNH, Huynh HH, Huynh PK, Le NQK. An in-depth review of AI-powered advancements in cancer drug discovery. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167680. [PMID: 39837431 DOI: 10.1016/j.bbadis.2025.167680] [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: 10/18/2024] [Revised: 01/12/2025] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
The convergence of artificial intelligence (AI) and genomics is redefining cancer drug discovery by facilitating the development of personalized and effective therapies. This review examines the transformative role of AI technologies, including deep learning and advanced data analytics, in accelerating key stages of the drug discovery process: target identification, drug design, clinical trial optimization, and drug response prediction. Cutting-edge tools such as DrugnomeAI and PandaOmics have made substantial contributions to therapeutic target identification, while AI's predictive capabilities are driving personalized treatment strategies. Additionally, advancements like AlphaFold highlight AI's capacity to address intricate challenges in drug development. However, the field faces significant challenges, including the management of large-scale genomic datasets and ethical concerns surrounding AI deployment in healthcare. This review underscores the promise of data-centric AI approaches and emphasizes the necessity of continued innovation and interdisciplinary collaboration. Together, AI and genomics are charting a path toward more precise, efficient, and transformative cancer therapeutics.
Collapse
Affiliation(s)
- Minh Huu Nhat Le
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; AIBioMed Research Group, Taipei Medical University, Taipei 110, Taiwan
| | - Phat Ky Nguyen
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; AIBioMed Research Group, Taipei Medical University, Taipei 110, Taiwan.
| | | | - Hien Quang Nguyen
- Cardiovascular Research Department, Methodist Hospital, Merrillville, IN 46410, USA
| | - Dao Ngoc Hien Tam
- Regulatory Affairs Department, Asia Shine Trading & Service Co. LTD, Viet Nam
| | - Han Hong Huynh
- International Master Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Phat Kim Huynh
- Department of Industrial and Systems Engineering, North Carolina A&T State University, Greensboro, NC 27411, USA.
| | - Nguyen Quoc Khanh Le
- AIBioMed Research Group, Taipei Medical University, Taipei 110, Taiwan; In-Service Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan.
| |
Collapse
|
158
|
Gyebi GA, Ejoh JC, Ogunyemi OM, Afolabi SO, Ibrahim IM, Anyanwu GO, Olorundare OE, Adebayo JO, Koketsu M. Cholinergic Inhibition and Antioxidant Potential of Gongronema latifolium Benth Leaf in Neurodegeneration: Experimental and In Silico Study. Cell Biochem Biophys 2025; 83:1-23. [PMID: 39120857 DOI: 10.1007/s12013-024-01467-7] [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] [Accepted: 07/25/2024] [Indexed: 08/10/2024]
Abstract
The use of Gongronema latifolium for the management of various forms of neurological disorders has generated a lot of interest in the need to further investigate its neurotherapeutic constituents. This work, therefore, focused on assessing the inhibitory potential of selected bioactive components derived from G. latifolium against key neurotherapeutic targets and oxidant species associated with neurodegeneration using in vitro analysis and biomolecular modelling. G. latifolium methanol extract (GLME), solvent partition, chromatographic fractions (A-F) of GLME and pregnane compounds (Iloneoside and marsectohexol) derived from fraction-B with the highest activity were investigated for in vitro acetylcholinesterase (AChE), butyrylcholinesterase (BChE), and monoamine oxidase (MAO) inhibition in addition to their in vitro antioxidant activities. The interactions of iloneoside, marsectohexol, and reference drugs with human acetylcholinesterase, butyrylcholinesterase, and β-secretase (BACE-1) were further assessed using molecular docking, binding free energy calculations, cluster analysis, and molecular dynamics simulations. The GLME and fractions inhibited the activities of both acetylcholinesterase and butyrylcholinesterase in a dose-dependent manner. Iloneoside and marsectohexol exhibited in vitro concentration-dependent inhibitory activities against acetylcholinesterase (IC50 = 19.28, 184.9 µM, respectively) and butyrylcholinesterase (IC50 = 30.75, 43.4 µM, respectively). These compounds also possess ferric ion-reducing, hydroxyl, and superoxide radical-scavenging activities. Iloneoside had the highest docking scores of -9.8, -9.9 -9.4 Kcal for AChE, BChE, and BACE1, respectively. The stability of the interaction of the bioactive compounds with the catalytic residues of the protein targets was preserved in a 100 ns molecular dynamics simulation. Iloneoside, a rare pregnane glycoside, was identified as a neurotherapeutic constituent of G. latifolium leaf. Further studies are suggested to investigate the neurotherapeutic potential in animal models.
Collapse
Affiliation(s)
- Gideon A Gyebi
- Department of Biochemistry, Bingham University, Karu, Nigeria.
- Department of Biotechnology and Food Science, Durban University of Technology, Durban, 4000, South Africa.
| | - Joseph C Ejoh
- Department of Biochemistry, Bingham University, Karu, Nigeria
| | - Oludare M Ogunyemi
- Nutritional and Industrial Biochemistry Research Unit, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, 200005, Nigeria
| | - Saheed O Afolabi
- Department of Pharmacology and Therapeutics, University of Ilorin, Ilorin, Nigeria
| | | | | | | | - Joseph O Adebayo
- Department of Biochemistry, University of Ilorin, Ilorin, Nigeria
| | - Mamoru Koketsu
- Department of Chemistry and Biomolecular Science, Gifu University, Gifu, Japan
| |
Collapse
|
159
|
Cheung LKY, Thallmair S, Yada RY. Elucidating the structure and function of a membrane-active plant protein domain using in silico mutagenesis. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2025; 1867:184409. [PMID: 39788471 DOI: 10.1016/j.bbamem.2025.184409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 12/28/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
The Solanum tuberosum (common potato) plant specific insert (StPSI) is an antimicrobial protein domain that exhibits membrane-disrupting and membrane-fusing activity upon dimerization at acidic pH, activity proposed to involve electrostatic attraction and membrane anchoring mediated by specific positively-charged and conserved tryptophan residues, respectively. This study is the first to employ an in silico mutagenesis approach to clarify the structure-function relationship of a plant specific insert (PSI), where ten rationally-mutated StPSI variants were investigated using all-atom and coarse-grained molecular dynamics. The tryptophan (W) residue at position 18 (W18) of wild-type StPSI was predicted to confer structural flexibility to the dimer and mediate a partial separation of the assembled monomers upon bilayer contact, while residues including W77 and the lysine (K) residue at position 83 (K83) were predicted to stabilize secondary structure and influence association with the model membrane. Mechanisms predicted to influence StPSI-membrane association included the partial separation of assembled monomers on the bilayer surface, formation of a specific salt bridge, and membrane anchoring of hinge 2 residues. The findings suggested that the structure-function relationship of StPSI involved several mechanisms that may each be modulated by specific key residues, insights that may support efforts to develop PSI with tailored membrane association for novel applications in plant biotechnology and crop protection.
Collapse
Affiliation(s)
- Lennie K Y Cheung
- Land and Food Systems, University of British Columbia, Vancouver, Canada
| | | | - Rickey Y Yada
- Land and Food Systems, University of British Columbia, Vancouver, Canada; Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, Canada.
| |
Collapse
|
160
|
Hu Z, Martí J. Unraveling atomic-scale mechanisms of GDP extraction catalyzed by SOS1 in KRAS-G12 and KRAS-D12 oncogenes. Comput Biol Med 2025; 186:109599. [PMID: 39731920 DOI: 10.1016/j.compbiomed.2024.109599] [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: 10/02/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/30/2024]
Abstract
The guanine exchange factor SOS1 plays a pivotal role in the positive feedback regulation of the KRAS signaling pathway. Recently, the regulation of KRAS-SOS1 interactions and KRAS downstream effector proteins has emerged as a key focus in the development of therapies targeting KRAS-driven cancers. However, the detailed dynamic mechanisms underlying SOS1-catalyzed GDP extraction and the impact of KRAS mutations remain largely unexplored. In this study, we unveil and describe in atomic detail the primary mechanisms by which SOS1 facilitates GDP extraction from KRAS oncogenes. For GDP-bound wild-type KRAS (KRAS-G12), four critical amino acids (Lys811, Glu812, Lys939, and Glu942) are identified as essential for the catalytic function of SOS1. Notably, the KRAS-G12D mutation (KRAS-D12) significantly accelerates the rate of GDP extraction. The molecular basis of this enhancement are attributed to hydrogen bonding interactions between the mutant residue Asp12 and a positively charged pocket in the intrinsically disordered region (residues 807-818), comprising Ser807, Trp809, Thr810, and Lys811. These findings provide novel insights into SOS1-KRAS interactions and offer a foundation for developing anti-cancer strategies aimed at disrupting these mechanisms.
Collapse
Affiliation(s)
- Zheyao Hu
- Department of Physics, Polytechnic University of Catalonia-Barcelona Tech, B4-B5 Northern Campus UPC, Barcelona, 08034, Catalonia, Spain
| | - Jordi Martí
- Department of Physics, Polytechnic University of Catalonia-Barcelona Tech, B4-B5 Northern Campus UPC, Barcelona, 08034, Catalonia, Spain.
| |
Collapse
|
161
|
Gyebi GA, Afolabi SO, Ogunyemi OM, Ibrahim IM, Olorundare OE, Adebayo JO, Koketsu M. Apoptotic Potential of Iloneoside from Gongronema latifolium Benth against Prostate Cancer Cells Using In Vitro and In Silico Approach. Cell Biochem Biophys 2025; 83:755-776. [PMID: 39302620 PMCID: PMC11870900 DOI: 10.1007/s12013-024-01507-2] [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] [Accepted: 08/25/2024] [Indexed: 09/22/2024]
Abstract
Prostate cancer is a major cause of cancer-related mortality in men worldwide. The anti-proliferative activity of Gongronema latifolium leaf extracts on some cancer cells has been reported. Herein, we investigated the growth inhibitory effect of the Gongronema latilolium leaf methanol extract and isolated pregnane (iloneoside) against prostate cancer cell lines using the MTT cell proliferation assay, apoptosis quantification, cell cycle analysis using flow cytometry and computational analysis molecular docking, molecular dynamics simulation (MDs), binding free energy computation and cluster analysis. In addition, UPLC-ESI-TOFMS chemical fingerprinting of previously isolated compounds was performed. The extract inhibited the growth of the cell lines with an IC50 of 49.3 µg/ml and 28.4 µg/ml for 24 h and 48 h, respectively, for PC3; and 43.7 µg/ml and 22.3 µg/ml for 24 h and 48 h, respectively, for DU145. Iloneoside demonstrated low inhibitory activities against PC3 and DU145 (IC50 > 80 μM). Apoptotic quantification and cell cycle analysis further showed that iloneoside induced apoptosis in a few cells at a dose of 200 uM. The ensemble-based molecular docking of the iloneoside to BCL-XL and BCL-2 proteins, and docking to MCL-1, BCL-A1 and BFL-1 proteins, respectively, presented binding energies of -7.22 ± 0.5, -8.12 ± 0.55, -7.1, -7.2 and -6.3 kcal/mol, while the MM/PBSA binding free energy was -25.72 ± 7.22 and -27.76 ± 11.32 kcal/mol for BCL-XL and BCL-2 proteins. Furthermore, iloneoside was stable during the 100 ns MDs analysis, while the clustering of the MDs trajectories showed that the interactions were strongly preserved. Iloneoside, in part, or in synergy with other constituents, may be responsible for the antiproliferative activities of the leaf, subject to further investigation.
Collapse
Affiliation(s)
- Gideon A Gyebi
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban, South Africa.
- Natural Products and Structural (Bio-Chem)-informatics Research Laboratory (NpsBC-RL), Department of Biochemistry, Faculty of Science and Technology, Bingham University, Karu, Nigeria.
| | - Saheed O Afolabi
- Biomolecular Modeling and Nutraceuticals Laboratory, Nutritional and Industrial Biochemistry Research Unit, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Oludare M Ogunyemi
- Faculty of Basic Medical Sciences, Department of Pharmacology and Therapeutics, University of Ilorin, Ilorin, Nigeria
| | - Ibrahim M Ibrahim
- Department of Biophysics, Faculty of Sciences, Cairo University, Giza, Egypt
| | - Olufunke E Olorundare
- Faculty of Basic Medical Sciences, Department of Pharmacology and Therapeutics, University of Ilorin, Ilorin, Nigeria
| | - Joseph O Adebayo
- Department of Biochemistry, Faculty of Life Sciences, University of Ilorin, Ilorin, Nigeria
| | - Mamoru Koketsu
- Faculty of Engineering, Department of Chemistry and Biomolecular Science, Gifu University, Gifu, Japan
| |
Collapse
|
162
|
Cui S, Dasgupta S, Yagi S, Kimura M, Furukawa R, Tagami S, Akanuma S. Insights into the low-temperature adaptation of an enzyme as studied through ancestral sequence reconstruction. Protein Sci 2025; 34:e70071. [PMID: 39968914 PMCID: PMC11836894 DOI: 10.1002/pro.70071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/17/2025] [Accepted: 02/04/2025] [Indexed: 02/20/2025]
Abstract
For billions of years, enzymes have evolved in response to the changing environments in which their host organisms lived. Various lines of evidence suggest the earliest primitive organisms inhabited high-temperature environments and possessed enzymes adapted to such conditions. Consequently, extant mesophilic and psychrophilic enzymes are believed to have adapted to lower temperatures during the evolutionary process. Herein, we analyzed this low-temperature adaptation using ancestral sequence reconstruction. Previously, we generated the phylogenetic tree of 3-isopropylmalate dehydrogenases (IPMDHs) and reconstructed the sequence of the last bacterial common ancestor. The corresponding ancestral enzyme displayed high thermostability and catalytic activity at elevated temperatures but moderate activity at low temperatures (Furukawa et al., Sci. Rep., 2020;10:15493). Here, to identify amino acid residues that are responsible for the low-temperature adaptation, we reconstructed and characterized all 11 evolutionary intermediates that sequentially connect the last bacterial common ancestor with extant mesophilic IPMDH from Escherichia coli. A remarkable change in catalytic properties, from those suited for high reaction temperatures to those adapted for low temperatures, occurred between two consecutive evolutionary intermediates. Using a combination of sequence comparisons between ancestral proteins and site-directed mutagenesis analyses, three key amino acid substitutions were identified that enhance low-temperature catalytic activity. Intriguingly, amino acid substitutions that had the most significant impact on activity at low temperatures displayed no discernable effect on thermostability. However, these substitutions markedly reduced the activation energy for catalysis, thereby improving low-temperature activity. The results were further investigated by molecular dynamics simulations of the predicted structures of the ancestral enzymes. Our findings exemplify how ancestral sequence reconstruction can identify residues crucial for adaptation to low temperatures.
Collapse
Affiliation(s)
- Shuang Cui
- Faculty of Human SciencesWaseda UniversityTokorozawaJapan
| | | | - Sota Yagi
- Faculty of Human SciencesWaseda UniversityTokorozawaJapan
- RIKEN Center for Biosystems Dynamics ResearchYokohamaJapan
| | - Madoka Kimura
- Faculty of Human SciencesWaseda UniversityTokorozawaJapan
| | | | | | | |
Collapse
|
163
|
Canini G, Mazzinelli E, Nocca G, Lattanzi W, Arcovito A. Targeting Glucosylceramide Synthase: Innovative Drug Repurposing Strategies for Lysosomal Diseases. Int J Mol Sci 2025; 26:2195. [PMID: 40076817 PMCID: PMC11900012 DOI: 10.3390/ijms26052195] [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: 01/13/2025] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Sphingolipidoses, a subgroup of lysosomal storage diseases (LSDs), are rare and debilitating disorders caused by defects in sphingolipid metabolism. Despite advancements in treatment, therapeutic options remain limited. Miglustat, a glucosylceramide synthase EC 2.4.1.80 (GCS) inhibitor, is one of the few available pharmacological treatments; however, it is associated with significant adverse effects that impact patients' quality of life. Drug repurposing offers a promising strategy to identify new therapeutic agents from approved drugs, expanding treatment options for rare diseases with limited therapeutic alternatives. This study aims to identify potential alternative inhibitors of GCS through a drug-repurposing approach, using computational and experimental methods to assess their therapeutic potential for sphingolipidoses. A library of approved drugs was screened using advanced computational techniques, including molecular docking, molecular dynamics simulations, and metadynamics, to identify potential GCS inhibitors. Promising candidates were selected for further in vitro validation to evaluate their inhibitory activity and potential as therapeutic alternatives to Miglustat. Computational screening identified several potential GCS inhibitors, with Dapagliflozin emerging as the most promising candidate. Experimental validation confirmed its efficacy, revealing a complementary mechanism of action to Miglustat while potentially offering a more favorable side effect profile. This study underscores the utility of computational and experimental methodologies in drug repurposing for rare diseases. The identification of Dapagliflozin as a potential GCS inhibitor provides a foundation for further preclinical and clinical evaluation, supporting its potential application in the treatment of sphingolipidoses.
Collapse
Affiliation(s)
- Giorgia Canini
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (G.C.); (E.M.); (G.N.)
| | - Elena Mazzinelli
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (G.C.); (E.M.); (G.N.)
| | - Giuseppina Nocca
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (G.C.); (E.M.); (G.N.)
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy;
| | - Wanda Lattanzi
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy;
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Alessandro Arcovito
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy; (G.C.); (E.M.); (G.N.)
- Fondazione Policlinico Universitario “A. Gemelli”, IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy;
| |
Collapse
|
164
|
Catte A, Oganesyan VS. Predicting and interpreting EPR spectra of POPC lipid bilayers with transmembrane α-helical peptides from all-atom molecular dynamics simulations. Phys Chem Chem Phys 2025; 27:4775-4784. [PMID: 39950932 DOI: 10.1039/d4cp04802d] [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: 02/28/2025]
Abstract
This study reports a large-scale all-atom MD simulation of POPC lipid bilayers in the presence of different concentrations of the transmembrane peptide acetyl-K2(LA)12K2-amide ((LA)12) and doped with 5-PC paramagnetic spin probes used in EPR studies. We apply a combined MD-EPR simulation methodology for the prediction of EPR spectra directly and completely from MD trajectories. This approach serves three major purposes. Firstly, comparing predicted EPR spectra with experimental ones, which are highly sensitive to motions, provides an ultimate test bed for the force fields currently employed for modeling lipid bilayer systems with embedded proteins or peptides. Secondly, simulations of EPR spectra directly from the atomistic MD models simplify the interpretation of the EPR line shapes and their changes induced by the presence of peptides in the lipid bilayer. These changes are directly linked to the dynamics and order of spin probes and POPC host molecules. Lastly and importantly, we demonstrate how the MD-EPR methodology can be employed to test the validity and limitations of the widely used approach for the estimation of the order parameter of lipids directly from the EPR experimental line shapes.
Collapse
Affiliation(s)
- Andrea Catte
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich, NR4 7TJ, UK.
| | - Vasily S Oganesyan
- School of Chemistry, Pharmacy and Pharmacology, University of East Anglia, Norwich, NR4 7TJ, UK.
| |
Collapse
|
165
|
Angelo M, Zhang W, Vilseck J, Aoki S. In silico λ-dynamics predicts protein binding specificities to modified RNAs. Nucleic Acids Res 2025; 53:gkaf166. [PMID: 40066880 PMCID: PMC11894534 DOI: 10.1093/nar/gkaf166] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/15/2025] Open
Abstract
RNA modifications shape gene expression through a variety of chemical changes to canonical RNA bases. Although numbering in the hundreds, only a few RNA modifications are well characterized, in part due to the absence of methods to identify modification sites. Antibodies remain a common tool to identify modified RNA and infer modification sites through straightforward applications. However, specificity issues can result in off-target binding and confound conclusions. This work utilizes in silico λ-dynamics to efficiently estimate binding free energy differences of modification-targeting antibodies between a variety of naturally occurring RNA modifications. Crystal structures of inosine and N6-methyladenosine (m6A) targeting antibodies bound to their modified ribonucleosides were determined and served as structural starting points. λ-Dynamics was utilized to predict RNA modifications that permit or inhibit binding to these antibodies. In vitro RNA-antibody binding assays supported the accuracy of these in silico results. High agreement between experimental and computed binding propensities demonstrated that λ-dynamics can serve as a predictive screen for antibody specificity against libraries of RNA modifications. More importantly, this strategy is an innovative way to elucidate how hundreds of known RNA modifications interact with biological molecules without the limitations imposed by in vitro or in vivo methodologies.
Collapse
Affiliation(s)
- Murphy Angelo
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202, United States
| | - Wen Zhang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202, United States
- Melvin and Bren Simon Cancer Center, 535 Barnhill Drive, Indianapolis, IN 46202, United States
| | - Jonah Z Vilseck
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202, United States
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Scott T Aoki
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, 635 Barnhill Drive, Indianapolis, IN 46202, United States
- Melvin and Bren Simon Cancer Center, 535 Barnhill Drive, Indianapolis, IN 46202, United States
| |
Collapse
|
166
|
Romeo E, Saccoliti F, Ocello R, Andonaia A, Allegretta C, Pastorino C, Pedemonte N, Falchi F, Laselva O, Bandiera T, Bertozzi F. Target Identification with Live-Cell Photoaffinity Labeling and Mechanism of Action Elucidation of ARN23765, a Highly Potent CFTR Corrector. J Med Chem 2025; 68:4596-4618. [PMID: 39928576 PMCID: PMC11873939 DOI: 10.1021/acs.jmedchem.4c02654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/12/2025]
Abstract
Molecular-targeted therapies for the treatment of cystic fibrosis (CF) rely on small-molecule modulators that rescue the activity of the defective CF transmembrane conductance regulator (CFTR) anion channel. ARN23765 is a small molecule with subnanomolar potency in rescuing the function of mutant CFTR in bronchial epithelial cells from CF patients carrying the F508del-CFTR mutation. Considering the multifaceted interactions of CFTR with the plasma membrane and the complexity of the protein network within the cellular compartments, here we report the investigation of ARN23765's molecular mechanism in live cells. We used the photoaffinity labeling (PAL) approach to demonstrate the interaction of ARN23765-derived probes with CFTR in cells. We showed that ARN23765 contributes to F508del-CFTR rescue by stabilizing the membrane-spanning domain-1 and interacting with CFTR at the same site as other type I CFTR correctors. Our study characterizes ARN23765's mode of action and highlights the potential of studying the interactions between CFTR and its correctors in live cells.
Collapse
Affiliation(s)
- Elisa Romeo
- Structural
Biophysics Facility, Istituto Italiano di
Tecnologia (IIT), Genova 16163, Italy
| | - Francesco Saccoliti
- D3-PharmaChemistry, Istituto Italiano di Tecnologia (IIT), Genova 16163, Italy
| | - Riccardo Ocello
- Department
of Pharmacy and Biotechnology, University
of Bologna, Bologna 40126, Italy
- Computational
and Chemical Biology, Istituto Italiano
di Tecnologia (IIT), Genova 16163, Italy
| | - Angela Andonaia
- D3-PharmaChemistry, Istituto Italiano di Tecnologia (IIT), Genova 16163, Italy
| | - Caterina Allegretta
- Department
of Clinical and Experimental Medicine, University
of Foggia, Foggia 71122, Italy
| | - Cristina Pastorino
- U.O.C.
Genetica
Medica, Istituto Giannina Gaslini (IGG), Genova 16147, Italy
| | - Nicoletta Pedemonte
- U.O.C.
Genetica
Medica, Istituto Giannina Gaslini (IGG), Genova 16147, Italy
| | - Federico Falchi
- Department
of Pharmacy and Biotechnology, University
of Bologna, Bologna 40126, Italy
- Computational
and Chemical Biology, Istituto Italiano
di Tecnologia (IIT), Genova 16163, Italy
| | - Onofrio Laselva
- Department
of Clinical and Experimental Medicine, University
of Foggia, Foggia 71122, Italy
| | - Tiziano Bandiera
- D3-PharmaChemistry, Istituto Italiano di Tecnologia (IIT), Genova 16163, Italy
| | - Fabio Bertozzi
- D3-PharmaChemistry, Istituto Italiano di Tecnologia (IIT), Genova 16163, Italy
| |
Collapse
|
167
|
Aonsri C, Kuljarusnont S, Tungmunnithum D. Discovering Skin Anti-Aging Potentials of the Most Abundant Flavone Phytochemical Compound Reported in Siam Violet Pearl, a Medicinal Plant from Thailand by In Silico and In Vitro Assessments. Antioxidants (Basel) 2025; 14:272. [PMID: 40227229 PMCID: PMC11939551 DOI: 10.3390/antiox14030272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 04/15/2025] Open
Abstract
Currently, nutraceuticals and functional food/cosmeceutical sectors are seeking natural molecules to develop various types of phytopharmaceutical products. Flavonoids have been reported in antioxidant and many medical/pharmacological activities. Monochoria angustifolia or Siam violet pearl medicinal plant is the newest species of the genus Monochoria C. Presl, which have long been consumed as food and herbal medicines. Though previous work showed that apigenin-7-O-glucoside is the most abundant antioxidant phytochemical found in this medicinal plant, the report on anti-aging activity is still lacking and needs to be filled in. The objective of this work is to explore anti-aging capacities of the most abundant antioxidant phytochemical reported in this plant using both in silico and in vitro assessments. In addition, pharmacokinetic properties were predicted. Interestingly, the results from both in silico and in vitro analysis showed a similar trend that apigenin-7-O-glucoside is a potential anti-aging agent against three enzymes. The pharmacokinetic properties, such as adsorption, distribution, metabolism, excretion and toxicity (ADMET), of this compound are also provided in this work. The current study is also the first report on anti-aging properties of this Thai medicinal plant. However, the safety and efficacy of future developed products from this compound and clinical study should be determined in the future.
Collapse
Affiliation(s)
- Chaiyawat Aonsri
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand;
- Unit of Compounds Library for Drug Discovery, Mahidol University, Bangkok 10400, Thailand
| | - Sompop Kuljarusnont
- Department of Obstetrics and Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand;
| | - Duangjai Tungmunnithum
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
- Le Studium Institute for Advanced Studies, 1 Rue Dupanloup, 45000 Orléans, France
| |
Collapse
|
168
|
Riepl D, Abou-Hamdan A, Gellner J, Biner O, Sjöstrand D, Högbom M, von Ballmoos C, Kaila VRI. Molecular Principles of Proton-Coupled Quinone Reduction in the Membrane-Bound Superoxide Oxidase. J Am Chem Soc 2025; 147:6866-6879. [PMID: 39937715 PMCID: PMC11869295 DOI: 10.1021/jacs.4c17055] [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: 11/30/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/14/2025]
Abstract
Reactive oxygen species (ROS) are physiologically harmful radical species generated as byproducts of aerobic respiration. To detoxify ROS, most cells employ superoxide scavenging enzymes that disproportionate superoxide (O2·-) to oxygen (O2) and hydrogen peroxide (H2O2). In contrast, the membrane-bound superoxide oxidase (SOO) is a minimal 4-helical bundle protein that catalyzes the direct oxidation of O2·- to O2 and drives quinone reduction by mechanistic principles that remain unknown. Here, we combine multiscale hybrid quantum/classical (QM/MM) free energy calculations and microsecond molecular dynamics simulations with functional assays and site-directed mutagenesis experiments to probe the mechanistic principles underlying the charge transfer reactions of the superoxide-driven quinone reduction. We characterize a cluster of charged residues at the periplasmic side of the membrane that functions as a O2·- collecting antenna, initiating electron transfer via two b hemes to the active site for quinone reduction at the cytoplasmic side. Based on multidimensional QM/MM string simulations, we find that a proton-coupled electron transfer (PCET) reaction from the active site heme b and nearby histidine residues (H87, H158) results in quinol (QH2) formation, followed by proton uptake from the cytoplasmic side of the membrane. The functional relevance of the identified residues is supported by site-directed mutagenesis and activity assays, with mutations leading to inhibition of the O2·--driven quinone reduction activity. We suggest that the charge transfer reactions could build up a proton motive force that supports the bacterial energy transduction machinery, while the PCET machinery provides unique design principles of a minimal oxidoreductase.
Collapse
Affiliation(s)
- Daniel Riepl
- Department
of Biochemistry and Biophysics, The Arrhenius Laboratories for Natural
Sciences, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Abbas Abou-Hamdan
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, CH-3012 Bern, Switzerland
| | - Jonas Gellner
- Department
of Biochemistry and Biophysics, The Arrhenius Laboratories for Natural
Sciences, Stockholm University, SE-106 91 Stockholm, Sweden
- Department
of Chemistry, Technical University Munich, D-85748 Garching, Germany
| | - Olivier Biner
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, CH-3012 Bern, Switzerland
| | - Dan Sjöstrand
- Department
of Biochemistry and Biophysics, The Arrhenius Laboratories for Natural
Sciences, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Martin Högbom
- Department
of Biochemistry and Biophysics, The Arrhenius Laboratories for Natural
Sciences, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Christoph von Ballmoos
- Department
of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, CH-3012 Bern, Switzerland
| | - Ville R. I. Kaila
- Department
of Biochemistry and Biophysics, The Arrhenius Laboratories for Natural
Sciences, Stockholm University, SE-106 91 Stockholm, Sweden
| |
Collapse
|
169
|
Reheim MAMA, Rady HSA, Mohamed OA, Hassan A, Hafiz ISA, Reffat HM, Elsaid FG, Eldesoqui M, Alshaya DS, Badawy AA, Fayad E, Abdelmonsef AH. Synthesis, Anti-Inflammatory, and Molecular Docking Studies of New Heterocyclic Derivatives Comprising Pyrazole, Pyridine, and/or Pyran Moieties. Pharmaceuticals (Basel) 2025; 18:335. [PMID: 40143114 PMCID: PMC11944836 DOI: 10.3390/ph18030335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
Introduction: Inhibiting cyclooxygenase-2 (COX-2) is a potential strategy in inflammation therapy. Thus, developing COX-2 inhibitors plays a pivotal role in efficient inflammation treatment. This study discloses the synthesis of new heterocyclic compounds incorporating pyridine, pyran, and/or pyrazole moieties as COX-2 inhibitors. Methods: In this study, the Claisen-Schmidt reaction of 1-(5-hydroxy-1,3-diphenyl-1H-pyrazol-4-yl)ethan-1-one 1 and p-methoxybenzaldehyde in ethanol containing aqueous sodium hydroxide (10%) led to the formation of 1-(5-hydroxy-1,3-diphenyl-1H-pyrazol-4-yl)-3-(4-methoxyphenyl)prop-2-en-1-one) 2. The latter compound was allowed to react as a key precursor with various nucleophiles such as ethyl cyanoacetate, malononitrile, cyclohexanone, ethyl acetoacetate, hydrazine, cyano acid hydrazide, hydrazide, and/or thiosemicarbazide to yield new heterocyclic derivatives comprising pyridine, pyran, and/or pyrazole moieties 3-15, according to the Michael addition reaction. The newly synthesized compounds were depicted using spectroscopic techniques such as IR, 1H-NMR, 13C-NMR, and MS. Moreover, their anti-inflammatory efficiency was in vitro evaluated by means of protein denaturation inhibition and cell membrane protection assay. Results: The results of 2-ΔΔct values of COX-2 expression for compounds 6, 11, 12, and 13 were 6.6, 2.9, 25.8, and 10.1, respectively. Therefore, compound 12, followed by 13, 11, and 6, showed potent anti-inflammatory properties by in vitro evaluation. Further, an in silico molecular docking study was performed on the best-docked compounds and reference drug (Diclofenac) to investigate their binding affinities against the active site of the target enzyme. The obtained results from the in silico study aligned with the biological evaluation. Conclusions: The studies open new doors for designing new heterocycles containing pyridine, pyran, and/or pyrazole moieties as potent anti-inflammatory agents.
Collapse
Affiliation(s)
- Mohamed A. M. Abdel Reheim
- Department of Chemistry, Faculty of Science, Arish University, Arish 45511, Egypt; (M.A.M.A.R.); (H.S.A.R.); (I.S.A.H.); (H.M.R.)
| | - Hend S. Abdel Rady
- Department of Chemistry, Faculty of Science, Arish University, Arish 45511, Egypt; (M.A.M.A.R.); (H.S.A.R.); (I.S.A.H.); (H.M.R.)
| | - Omnia A. Mohamed
- Department of Biochemistry and Molecular Biology, Theodor Bilharz Research Institute, Giza 12411, Egypt;
| | - Abdelfattah Hassan
- Department of Medicinal Chemistry, Faculty of Pharmacy, South Valley University, Qena 83523, Egypt;
| | - Ibrahim S. Abdel Hafiz
- Department of Chemistry, Faculty of Science, Arish University, Arish 45511, Egypt; (M.A.M.A.R.); (H.S.A.R.); (I.S.A.H.); (H.M.R.)
| | - Hala M. Reffat
- Department of Chemistry, Faculty of Science, Arish University, Arish 45511, Egypt; (M.A.M.A.R.); (H.S.A.R.); (I.S.A.H.); (H.M.R.)
| | - Fahmy Gad Elsaid
- Department of Biology, College of Science, King Khalid University, P.O. Box 960, Abha 61421, Saudi Arabia;
| | - Mamdouh Eldesoqui
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia;
| | - Dalal Sulaiman Alshaya
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia;
| | - Abdelnaser A. Badawy
- Department of Biochemistry, Faculty of Medicine, Northern Border University, P.O. Box 1321, Arar 91431, Saudi Arabia;
| | - Eman Fayad
- Department of Biotechnology, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | | |
Collapse
|
170
|
Wu X, Brooks BR. Self-Guided Molecular Simulation to Enhance Concerted Motion. Int J Mol Sci 2025; 26:1969. [PMID: 40076595 PMCID: PMC11899740 DOI: 10.3390/ijms26051969] [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: 01/24/2025] [Revised: 02/18/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Self-guided (SG) molecular simulation methods, namely self-guided molecular dynamics (SGMD) and self-guided Langevin dynamics (SGLD), enhance conformational search by promoting low-frequency motion. A simple local time averaging scheme is used to extract low-frequency properties with little overhead in computing costs. For molecular processes to form ordered structures like ligand binding and protein folding, it is believed that concerted motions play crucial roles. To enhance the concerted motion in molecular systems, we propose a spatial averaging scheme to extract the concerted motion of a local region. Applying guiding forces based on spatial averaging, self-guided molecular simulations can enhance concerted motion and reach ordered structures more efficiently. Through simulations of amyloid fibril peptides, we demonstrated that the spatial averaging in self-guided Langevin dynamics results in accelerated β-sheet formation.
Collapse
Affiliation(s)
- Xiongwu Wu
- Laboratory of Computational Biology, National Heart, Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | | |
Collapse
|
171
|
Zaręba P, Drabczyk AK, Wnorowski A, Maj M, Rurka P, Malarz K, Latacz G, Nędza K, Ciura K, Greber KE, Boguszewska-Czubara A, Śliwa P, Kuliś J. Long-Chain Cyclic Arylguanidines as Multifunctional Serotonin Receptor Ligands with Antiproliferative Activity. ACS OMEGA 2025; 10:6446-6469. [PMID: 40028084 PMCID: PMC11866022 DOI: 10.1021/acsomega.4c06456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 03/05/2025]
Abstract
Recent investigations have shown serotonin's stimulatory effect on several types of cancers and carcinoid tumors. Nowadays there has been a significant increase in interest in 5-HT7 and 5-HT5A receptors in the context of cancer treatment. The possible role of 5-HT6R in the pathogenesis and progression of glioma remains an interesting and relatively unexplored issue. We developed a new group of long-chain 2-aminoquinazoline sulfonamides as new multifunctional serotonin receptor ligands, focused on 5-HT6R. The chosen group was further evaluated for antiproliferative effects on 1321N1 astrocytoma cells, along with U87MG, U-251, and LN-229 glioblastoma cell lines. Certain compounds were subjected to in vitro absorption, distribution, metabolism, excretion, and toxicity (ADMET) testing, for assessing factors such as lipophilicity, plasma protein binding, phospholipid affinity, potential for drug-drug interactions (DDI), membrane permeability (PAMPA), metabolic stability, and hepatotoxicity. Additionally, in vivo testing was performed using the Danio rerio model. The developed group includes the selective 5-HT6R antagonist PP 15, dual ligand for 5-HT1AR/5-HT6R PP 13, and dual ligand for 5-HT5AR/5-HT6R PP 10. The use of multifunctional ligands was associated with high anticancer activity both against selected glioma cell lines and other cancers (IC50 < 25 μM).
Collapse
Affiliation(s)
- Przemysław Zaręba
- Faculty
of Chemical Engineering and Technology, Department of Chemical Technology
and Environmental Analytics, Cracow University
of Technology, 24 Warszawska Street, 31-155 Cracow, Poland
| | - Anna K. Drabczyk
- Faculty
of Chemical Engineering and Technology, Department of Organic Chemistry
and Technology, Cracow University of Technology, 24 Warszawska Street, 31-155 Cracow, Poland
| | - Artur Wnorowski
- Department
of Biopharmacy, Medical University of Lublin, 4a Chodźki Street, 20-093 Lublin, Poland
| | - Maciej Maj
- Department
of Biopharmacy, Medical University of Lublin, 4a Chodźki Street, 20-093 Lublin, Poland
| | - Patryk Rurka
- Institute
of Physics, University of Silesia in Katowice, 1A 75 Pułku Piechoty Street, 41-500 Chorzow, Poland
| | - Katarzyna Malarz
- Institute
of Physics, University of Silesia in Katowice, 1A 75 Pułku Piechoty Street, 41-500 Chorzow, Poland
- Department
of Systems Biology and Engineering, Silesian
University of Technology, 11 Akademicka Street, 44-100 Gliwice, Poland
| | - Gniewomir Latacz
- Department
of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Cracow, Poland
| | - Krystyna Nędza
- Department
of Medicinal Chemistry, Maj Institute of
Pharmacology − Polish Academy of Sciences, 12 Smętna Street, 31-343 Cracow, Poland
| | - Krzesimir Ciura
- Department
of Physical Chemistry, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdansk, Poland
- Laboratory
of Environmental Chemoinformatics, Faculty of Chemistry, University of Gdansk, 63 Wita Stwosza Street, 80-308 Gdansk, Poland
| | - Katarzyna Ewa Greber
- Department
of Physical Chemistry, Faculty of Pharmacy, Medical University of Gdansk, 80-416 Gdansk, Poland
| | - Anna Boguszewska-Czubara
- Department
of Medical Chemistry, Medical University
of Lublin, 4a Chodźki
Street, 20-093 Lublin, Poland
| | - Paweł Śliwa
- Faculty
of Chemical Engineering and Technology, Department of Organic Chemistry
and Technology, Cracow University of Technology, 24 Warszawska Street, 31-155 Cracow, Poland
| | - Julia Kuliś
- Faculty
of Chemical Engineering and Technology, Department of Chemical Technology
and Environmental Analytics, Cracow University
of Technology, 24 Warszawska Street, 31-155 Cracow, Poland
| |
Collapse
|
172
|
Suh D, Schwartz R, Gupta PK, Zev S, Major DT, Im W. CHARMM-GUI EnzyDocker for Protein-Ligand Docking of Multiple Reactive States along a Reaction Coordinate in Enzymes. J Chem Theory Comput 2025; 21:2118-2128. [PMID: 39950957 PMCID: PMC11866752 DOI: 10.1021/acs.jctc.4c01691] [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: 12/10/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025]
Abstract
Enzymes play crucial roles in all biological systems by catalyzing a myriad of chemical reactions. These reactions range from simple one-step processes to intricate multistep cascades. Predicting mechanistically appropriate binding modes along a reaction pathway for substrate, product, and all reaction intermediates and transition states is a daunting task. To address this challenge, special docking programs like EnzyDock have been developed. Yet, running such docking simulations is complicated due to the nature of multistep enzyme processes. This work presents CHARMM-GUI EnzyDocker, a web-based cyberinfrastructure designed to streamline the preparation and running of EnzyDock docking simulations. The development of EnzyDocker has been achieved through integration of existing CHARMM-GUI modules, such as PDB Reader and Manipulator, Ligand Designer, and QM/MM Interfacer. In addition, new functionalities have been developed to facilitate a one-stop preparation of multistate and multiscale docking systems and enable interactive and intuitive ligand modifications and flexible protein residues selections. A simple setup related to multiligand docking is automatized through intuitive user interfaces. EnzyDocker offers support for standard classical docking and QM/MM docking with CHARMM built-in semiempirical engines. Automated consensus restraints for incorporating experimental knowledge into the docking are facilitated via a maximum common substructure algorithm. To illustrate the robustness of EnzyDocker, we conducted docking simulations of three enzyme systems: dihydrofolate reductase, SARS-CoV-2 Mpro, and the diterpene synthase CotB2. In addition, we have created four tutorial videos about these systems, which can be found at https://www.charmm-gui.org/demo/enzydock. EnzyDocker is expected to be a valuable and accessible web-based tool that simplifies and accelerates the setup process for multistate docking for enzymes.
Collapse
Affiliation(s)
- Donghyuk Suh
- Department
of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - 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
| | - Prashant Kumar Gupta
- Department
of Chemistry, Israel National Institute of Energy Storage (INIES)
and Institute for Nanotechnology & Advanced Materials, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Shani Zev
- Department
of Chemistry, Israel National Institute of Energy Storage (INIES)
and Institute for Nanotechnology & Advanced Materials, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - 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
| | - Wonpil Im
- Department
of Biological Sciences, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| |
Collapse
|
173
|
Alsenani F, Elmaidomy AH, Alaaeldin R, Abdel-Maqsoud NMR, Altemani FH, Algehainy NA, Alzubaidi MA, Bakhsh HT, Mokhtar FA, Elrehany MA, Sayed AM, Al-Sanea MM, Bringmann G, Abdelmohsen UR, Abbas GM. Olea europaea L. cv. Nepal Leaf Extract Alleviates Dyslipidemia and Inflammation Related to Obesity in Rabbits: A Network Pharmacology Approach Revealing Effective Antihyperlipidemic Bioactive Compounds. Chem Biodivers 2025:e202402110. [PMID: 40000423 DOI: 10.1002/cbdv.202402110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 02/12/2025] [Accepted: 02/25/2025] [Indexed: 02/27/2025]
Abstract
The current study investigated the effects of Olea europaea L. cv. Nepal (OEN) leaf extract on obesity-related disorders in rabbits. OEN extract, in dose 100 mg/kg body weight, significantly reduced cholesterol (TC), triglycerides (TAG), and inflammatory markers (MCP-1, VCAM-1, IL-6, IL-1β, TNFα, NF-κB) in both preventative and treatment groups. In addition, heart tissue displayed near-normal collagen fiber distribution. LC-HRESIMS analysis tentatively identified 20 metabolites (1-20) in OEN, including lignans, secoiridoids, and triterpenoids. Network pharmacology analysis suggested 8-hydroxy-p-menth-1-en-7-oic-acid (13), 6,7-dihydroxy-2H-1-benzopyran-2-one 6-O-d-glucopyranoside (5), and 3-hydroxy-12-oleanen-28-oic-acid (20) as key antihyperlipidemic compounds, potentially targeting pathways involved in hyperlipidemia regulation. Furthermore, in silico studies revealed that lignans (compounds 1, 2, and 16) from OEN may bind and inhibit HMG-CoA reductase, a crucial enzyme in cholesterol metabolism. These results indicate the potential of using OEN leaf extract as a therapeutic strategy for managing hyperlipidemia and inflammation associated with obesity. More investigation is necessary to confirm and validate these results and explore the extract's full therapeutic potential.
Collapse
Affiliation(s)
- Faisal Alsenani
- Department of Pharmaceutical Sciences, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Abeer H Elmaidomy
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Rania Alaaeldin
- Department of Biochemistry, Faculty of Pharmacy, Deraya University, New Minia, Egypt
| | | | - Faisal H Altemani
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Naseh A Algehainy
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Mubarak A Alzubaidi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Centre of Excellence in Bionanoscience, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hussain T Bakhsh
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Mahmoud A Elrehany
- Department of Biochemistry, Faculty of Pharmacy, Deraya University, New Minia, Egypt
| | - Ahmed M Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Nahda University, Beni-Suef, Egypt
| | - Mohammad M Al-Sanea
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
| | - Gerhard Bringmann
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, Würzburg, Germany
| | - Usama Ramadan Abdelmohsen
- Deraya Center for Scientific Research, Deraya University, New Minia, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Ghada M Abbas
- Department of Pharmacognosy, Faculty of Pharmacy, Horus University-Egypt (HUE, New Damietta, Egypt
| |
Collapse
|
174
|
Lazarenko D, Schmidt GP, Crowley MF, Beckham GT, Knott BC. Molecular Details of Polyester Decrystallization via Molecular Simulation. Macromolecules 2025; 58:1795-1803. [PMID: 40026450 PMCID: PMC11866931 DOI: 10.1021/acs.macromol.4c02130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 03/05/2025]
Abstract
Waste polyesters are a potential feedstock for recycled and upcycled products. These polymers are generally semicrystalline, which presents a challenge for chemical and biological recycling to monomers, and thus the thermodynamic work associated with polyester decrystallization is an important consideration in some depolymerization strategies. Here, we use molecular dynamics simulations to calculate the free energy required to decrystallize a single chain from the crystal surface of five commercially and scientifically important, semiaromatic polyesters (PET, PTT, PBT, PEN, and PEF) in water. Our results indicate the decrystallization work ranges from approximately 15 kcal/mol (PEN) to 8 kcal/mol (PEF) per repeat unit for chains in the middle of a crystal surface. The insight gained into the molecular interactions that form the structural basis of semicrystalline synthetic polyesters can guide the pursuit of more efficient plastic processing, which could include catalyst development, optimizing recycling conditions including pretreatment, enzyme and solvent selections, and design of new materials.
Collapse
Affiliation(s)
- Daria Lazarenko
- Renewable Resources and Enabling Sciences
Center, National Renewable Energy Laboratory, Golden, Colorado 80401, United States
| | - Graham P. Schmidt
- Renewable Resources and Enabling Sciences
Center, National Renewable Energy Laboratory, Golden, Colorado 80401, United States
| | - Michael F. Crowley
- Renewable Resources and Enabling Sciences
Center, National Renewable Energy Laboratory, Golden, Colorado 80401, United States
| | - Gregg T. Beckham
- Renewable Resources and Enabling Sciences
Center, National Renewable Energy Laboratory, Golden, Colorado 80401, United States
| | - Brandon C. Knott
- Renewable Resources and Enabling Sciences
Center, National Renewable Energy Laboratory, Golden, Colorado 80401, United States
| |
Collapse
|
175
|
Talagayev V, Chen Y, Doering NP, Obendorf L, Denzinger K, Puls K, Lam K, Liu S, Wolf CA, Noonan T, Breznik M, Knaus P, Wolber G. OpenMMDL - Simplifying the Complex: Building, Simulating, and Analyzing Protein-Ligand Systems in OpenMM. J Chem Inf Model 2025; 65:1967-1978. [PMID: 39933881 PMCID: PMC11863370 DOI: 10.1021/acs.jcim.4c02158] [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: 11/21/2024] [Revised: 01/21/2025] [Accepted: 01/30/2025] [Indexed: 02/13/2025]
Abstract
Molecular dynamics (MD) simulations have become an essential tool for studying the dynamics of biological systems and exploring protein-ligand interactions. OpenMM is a modern, open-source software toolkit designed for MD simulations. Until now, it has lacked a module dedicated to building receptor-ligand systems, which is highly useful for investigating protein-ligand interactions for drug discovery. We therefore introduce OpenMMDL, an open-source toolkit that enables the preparation and simulation of protein-ligand complexes in OpenMM, along with the subsequent analysis of protein-ligand interactions. OpenMMDL consists of three main components: OpenMMDL Setup, a graphical user interface based on Python Flask to prepare protein and simulation settings, OpenMMDL Simulation to perform MD simulations with consecutive trajectory postprocessing, and finally OpenMMDL Analysis to analyze simulation results with respect to ligand binding. OpenMMDL is not only a versatile tool for analyzing protein-ligand interactions and generating ligand binding modes throughout simulations; it also tracks and clusters water molecules, particularly those exhibiting minimal displacement from their previous coordinates, providing insights into solvent dynamics. We applied OpenMMDL to study ligand-receptor interactions across diverse biological systems, including LDN-193189 and LDN-212854 with ALK2 (kinases), nifedipine and amlodipine in Cav1.1 (ion channels), LSD in 5-HT2B (G-protein coupled receptors), letrozole in CYP19A1 (cytochrome P450 oxygenases), flavin mononucleotide binding the FMN-riboswitch (RNAs), ligand C08 bound to TLR8 (toll-like receptor), and PZM21 bound to MOR (opioid receptor), highlighting distinct functionalities of OpenMMDL. OpenMMDL is publicly available at https://github.com/wolberlab/OpenMMDL.
Collapse
Affiliation(s)
- Valerij Talagayev
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Yu Chen
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Niklas Piet Doering
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Leon Obendorf
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
- Department
of Biology, Chemistry and Pharmacy, Institute
of Biochemistry, Signal Transduction Group, Thielallee 64, 14195 Berlin, Germany
| | - Katrin Denzinger
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Kristina Puls
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Kevin Lam
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Sijie Liu
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Clemens Alexander Wolf
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Theresa Noonan
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Marko Breznik
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| | - Petra Knaus
- Department
of Biology, Chemistry and Pharmacy, Institute
of Biochemistry, Signal Transduction Group, Thielallee 64, 14195 Berlin, Germany
| | - Gerhard Wolber
- Department
of Biology, Chemistry and Pharmacy, Institute
of Pharmacy, Molecular Design Group, Königin-Luisestr. 2 + 4, 14195 Berlin, Germany
| |
Collapse
|
176
|
Fujisawa M, Onodera T, Kuroda D, Kewcharoenwong C, Sasaki M, Itakura Y, Yumoto K, Nithichanon A, Ito N, Takeoka S, Suzuki T, Sawa H, Lertmemongkolchai G, Takahashi Y. Molecular convergence of neutralizing antibodies in human revealed by repeated rabies vaccination. NPJ Vaccines 2025; 10:39. [PMID: 39988605 PMCID: PMC11847937 DOI: 10.1038/s41541-025-01073-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 01/13/2025] [Indexed: 02/25/2025] Open
Abstract
Rabies vaccines require repeated immunization to robustly elicit neutralizing antibodies that prevent fatal diseases. Here, we analyzed rabies glycoprotein antibody repertoires at both polyclonal and monoclonal levels following repeated vaccination. Booster vaccination dramatically elevated the neutralizing activity of recalled antibodies, primarily targeting an immunodominant site III epitope with hydrophilic and rugged structures. Strikingly, the majority of site III-directed antibodies in the recall response used a convergent VH gene (IGHV3-30), and they exhibited more hydrophilic and shorter paratopes than non-site III antibodies, providing physicochemical advantages for binding to site III. Additionally, several amino acids on heavy chain CDR3 were identified as key sites for acquiring an ultrapotent neutralizing activity through site III binding. Our in-depth analysis of antibody repertoires revealed the molecular signatures of neutralizing antibodies generated by repeated rabies vaccination, possibly as a result of adaptive convergence.
Collapse
Affiliation(s)
- Mizuki Fujisawa
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), Tokyo, Japan
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Taishi Onodera
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan.
| | - Daisuke Kuroda
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan.
| | - Chidchamai Kewcharoenwong
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Ching Mai, Thailand
- The Centre for Research & Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Michihito Sasaki
- Division of Molecular Pathobiology, International Institute for Zoonosis Control (IIZC), Hokkaido University, Hokkaido, Japan
- Institute for Vaccine Research and Development, Hokkaido University, Hokkaido, Japan
| | - Yukari Itakura
- Institute for Vaccine Research and Development, Hokkaido University, Hokkaido, Japan
| | - Kohei Yumoto
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Arnone Nithichanon
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Naoto Ito
- Laboratory of Zoonotic Diseases, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Shinji Takeoka
- Department of Life Science and Medical Bioscience, Graduate School of Advanced Science and Engineering, Waseda University (TWIns), Tokyo, Japan
- Research Institute for Science and Engineering, Waseda University, Tokyo, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hirofumi Sawa
- Institute for Vaccine Research and Development, Hokkaido University, Hokkaido, Japan
- One Health Research Center, Hokkaido University, Hokkaido, Japan
| | - Ganjana Lertmemongkolchai
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Ching Mai, Thailand
- The Centre for Research & Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan.
- Institute for Vaccine Research and Development, Hokkaido University, Hokkaido, Japan.
| |
Collapse
|
177
|
Seagle HM, Akerele AT, DeCorte JA, Hellwege JN, Breeyear JH, Kim J, Levin M, Khodurksy S, Bress A, Lee K, Meiler J, Gill D, Lee JS, Heberer K, Miller DR, Reaven P, Chang KM, Lynch JA, Khankari NK, Shuey MM, Edwards TL, Vujkovic M. Genomics-Informed Drug Repurposing Strategy Identifies Novel Therapeutic Targets for Metabolic Dysfunction-Associated Steatotic Liver Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.18.25321035. [PMID: 40034783 PMCID: PMC11875238 DOI: 10.1101/2025.02.18.25321035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Identification of drug-repurposing targets with genetic and biological support is an economically and temporally efficient strategy for improving treatment of diseases. We employed a cross-disciplinary approach to identify potential treatments for metabolic dysfunction associated steatotic liver disease (MASLD) using humans as a model organism. We identified 212 putative causal genes associated with MASLD using data from a large multi-ancestry genetic association study, of which 158 (74.5%) are novel. From this set we identified 57 genes that encode for druggable protein targets, and where the effects of increasing genetically predicted gene expression on MASLD risk align with the function of that drug on the protein target. These potential targets were then evaluated for evidence of efficacy using Mendelian randomization, pathway analysis, and protein structural modeling. Using these approaches, we present compelling evidence to suggest activation of FADS1 by icosopent ethyl as well as S1PR2 by fingolimod could be promising therapeutic strategies for MASLD.
Collapse
Affiliation(s)
- Hannah M Seagle
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Joseph Maxwell Cleland Atlanta VA Medical Center, Atlanta, Georgia, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Alexis T Akerele
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- School of Graduate Studies and Department of Microbiology, Immunology and Physiology, Meharry Medical College, Nashville, Tennessee, United States of America
- Division of Quantitative Science, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Joseph A DeCorte
- Vanderbilt Medical Scientist Training Program, Vanderbilt University Medical Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Chemical and Physical Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Jacklyn N Hellwege
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- VA Tennessee Valley Healthcare System (626), Nashville, Tennessee, United States of America
| | - Joseph H Breeyear
- Biostatistics and Computational Biology Branch, National Institute for Environmental Health Sciences, National Institutes of Health, Durham, North Carolina, United States of America
| | - Jeewoo Kim
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Division of Quantitative Science, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Medical Scientist Training Program, Vanderbilt University Medical Center, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Michael Levin
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Phoenix VA Health Care System; University of Arizona, Phoenix, Arizona, United States of
| | - Samuel Khodurksy
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Phoenix VA Health Care System; University of Arizona, Phoenix, Arizona, United States of
| | - Adam Bress
- Salt Lake City VA Medical Center, Salt Lake City, Utah, United States of America
- University of Utah, School of Medicine, Salt Lake City, Utah, United States of America
| | - Kyung Lee
- Salt Lake City VA Medical Center, Salt Lake City, Utah, United States of America
| | - Jens Meiler
- Department of Chemical and Physical Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Institute for Drug Discovery, Leipzig University Medical School, Leipzig, Germany
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Jennifer S Lee
- Stanford University, Stanford, California, United States of America
- Palo Alto VA Medical Center, Palo Alto, California, United States of America
| | - Kent Heberer
- Palo Alto VA Medical Center, Palo Alto, California, United States of America
| | - Donald R Miller
- VA Center for Medication Safety, Department of Veterans Affairs, Chicago, Illinois, United States of America
- Center for Population Health, Department of Biomedical and Nutritional Sciences, University of Massachusetts, Lowell, MA, United States of America
| | - Peter Reaven
- Phoenix VA Health Care System; University of Arizona, Phoenix, Arizona, United States of
| | - Kyong-Mi Chang
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Corporal Michael J. Crescenz Philadelphia VA Medical Center, Philadelphia, Pennsylvania, United States of America
| | - Julie A Lynch
- Salt Lake City VA Medical Center, Salt Lake City, Utah, United States of America
- University of Utah, School of Medicine, Salt Lake City, Utah, United States of America
| | - Nikhil K Khankari
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Megan M Shuey
- Vanderbilt University Genetics Institute, Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- VA Tennessee Valley Healthcare System (626), Nashville, Tennessee, United States of America
| | - Marijana Vujkovic
- University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
- Corporal Michael J. Crescenz Philadelphia VA Medical Center, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
178
|
Han Y, Dawson JR, DeMarco KR, Rouen KC, Ngo K, Bekker S, Yarov-Yarovoy V, Clancy CE, Xiang YK, Ahn SH, Vorobyov I. Molecular simulations reveal intricate coupling between agonist-bound β-adrenergic receptors and G protein. iScience 2025; 28:111741. [PMID: 39898043 PMCID: PMC11787599 DOI: 10.1016/j.isci.2024.111741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/24/2024] [Accepted: 12/18/2024] [Indexed: 02/04/2025] Open
Abstract
G protein-coupled receptors (GPCRs) and G proteins transmit signals from hormones and neurotransmitters across cell membranes, initiating downstream signaling and modulating cellular behavior. Using advanced computer modeling and simulation, we identified atomistic-level structural, dynamic, and energetic mechanisms of norepinephrine (NE) and stimulatory G protein (Gs) interactions with β-adrenergic receptors (βARs), crucial GPCRs for heart function regulation and major drug targets. Our analysis revealed distinct binding behaviors of NE within β1AR and β2AR despite identical orthosteric binding pockets. β2AR had an additional binding site, explaining variations in NE binding affinities. Simulations showed significant differences in NE dissociation pathways and receptor interactions with the Gs. β1AR binds Gs more strongly, while β2AR induces greater conformational changes in the α subunit of Gs. Furthermore, GTP and GDP binding to Gs may disrupt coupling between NE and βAR, as well as between βAR and Gs. These findings may aid in designing precise βAR-targeted drugs.
Collapse
Affiliation(s)
- Yanxiao Han
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
| | - John R.D. Dawson
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
- Biophysics Graduate Group, University of California, Davis, Davis, CA 95616, USA
| | - Kevin R. DeMarco
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
| | - Kyle C. Rouen
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
- Biophysics Graduate Group, University of California, Davis, Davis, CA 95616, USA
| | - Khoa Ngo
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
- Biophysics Graduate Group, University of California, Davis, Davis, CA 95616, USA
| | - Slava Bekker
- American River College, Sacramento, CA 95841, USA
| | - Vladimir Yarov-Yarovoy
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
- Department of Anesthesiology and Pain Medicine, University of California, Davis, Davis, CA 95616, USA
- Center for Precision Medicine and Data Science, University of California, Davis, Davis, CA 95616, USA
| | - Colleen E. Clancy
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
- Center for Precision Medicine and Data Science, University of California, Davis, Davis, CA 95616, USA
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Yang K. Xiang
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
- VA Northern California Health Care System, Mather, CA 95655, USA
| | - Surl-Hee Ahn
- Department of Chemical Engineering, University of California, Davis, Davis, CA 95616, USA
| | - Igor Vorobyov
- Department of Physiology and Membrane Biology, University of California, Davis, Davis, CA 95616, USA
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| |
Collapse
|
179
|
Joshi K, Miao Y. Mechanisms of Peptide Agonist Dissociation and Deactivation of Adhesion G-Protein-Coupled Receptors. Biochemistry 2025; 64:871-878. [PMID: 39902762 DOI: 10.1021/acs.biochem.4c00531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Adhesion G protein-coupled receptors (ADGRs) belong to Class B2 of GPCRs and are involved in a wide array of important physiological processes. ADGRs contain a GPCR autoproteolysis-inducing domain that is proximal to the receptor N-terminus and undergoes autoproteolysis during the biosynthesis to generate two fragments: the N-terminal fragment (NTF) and the C-terminal fragment (CTF). Dissociation of NTF reveals a tethered agonist to activate the CTF of ADGRs for G protein signaling. Synthetic peptides that mimic the tethered agonist can also activate ADGRs. However, mechanisms of peptide agonist dissociation and the deactivation of ADGRs remain poorly understood. In this study, we have performed all-atom enhanced sampling simulations using a novel protein-protein interaction Gaussian-accelerated molecular dynamics (PPI-GaMD) method on the ADGRG2-IP15 and ADGRG1-P7 complexes. The PPI-GaMD simulations captured the dissociation of the IP15 and P7 peptide agonists from their target receptors. We were able to identify important low-energy conformations of ADGRG2 and ADGRG1 in the active, intermediate, and inactive states, as well as different states of the peptide agonists IP15 and P7 during dissociation. Therefore, our PPI-GaMD simulations have revealed dynamic mechanisms of peptide agonist dissociation and deactivation of ADGRG1 and ADGRG2, which will facilitate the rational design of peptide regulators of the two receptors and other ADGRs.
Collapse
Affiliation(s)
- Keya Joshi
- Department of Pharmacology and Computational Medicine Program, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Yinglong Miao
- Department of Pharmacology and Computational Medicine Program, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
180
|
Kutzner C, Miletić V, Palacio Rodríguez K, Rampp M, Hummer G, de Groot BL, Grubmüller H. Scaling of the GROMACS Molecular Dynamics Code to 65k CPU Cores on an HPC Cluster. J Comput Chem 2025; 46:e70059. [PMID: 39945385 PMCID: PMC11822873 DOI: 10.1002/jcc.70059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025]
Abstract
We benchmarked the performance of the GROMACS 2024 molecular dynamics (MD) code on a modern high-performance computing (HPC) cluster with AMD CPUs on up to 65,536 CPU cores. We used five different MD systems, ranging in size from about 82,000 to 204 million atoms, and evaluated their performance using two different Message Passing Interface (MPI) libraries, Intel-MPI and Open-MPI. The largest system showed near-perfect strong scaling up to 512 nodes or 65,536 cores, maintaining a parallel efficiency above 0.9 even at the highest level of parallelization. Energy efficiency for a given number of nodes was generally equal to or slightly better than parallel efficiency. We achieved peak performances of 687 ns/d for the 82k atom system, 116 ns/d for the 53M atom system, and about 35 ns/d for the largest 204M atom system. These results demonstrate that highly optimized software running on a state-of-the-art HPC cluster provides sufficient computing power to simulate biomolecular systems at the mesoscale of viruses and organelles, and potentially small cells in the near future.
Collapse
Affiliation(s)
- Carsten Kutzner
- Theoretical and Computational BiophysicsMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | | | | | - Markus Rampp
- Max Planck Computing and Data FacilityGarchingGermany
| | - Gerhard Hummer
- Max Planck Institute of BiophysicsFrankfurt am MainGermany
| | - Bert L. de Groot
- Theoretical and Computational BiophysicsMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Helmut Grubmüller
- Theoretical and Computational BiophysicsMax Planck Institute for Multidisciplinary SciencesGöttingenGermany
| |
Collapse
|
181
|
Venable RM, Pane AJ, Rice A, Pastor RW. Effects of a Polarizable Force Field on Membrane Dynamics: Surface Viscosity, Lipid Diffusion, and Peptide Induced Pore Formation. J Comput Chem 2025; 46:e70001. [PMID: 39936400 PMCID: PMC11926808 DOI: 10.1002/jcc.70001] [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: 08/21/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 02/13/2025]
Abstract
The effects of the newly developed CHARMM polarizable lipid force field (FF), Drude2023, on selected lipid dynamical properties are compared with the additive CHARMM36 (C36), and an extension of C36, termed C36/LJ-PME, which includes long-range Lennard-Jones (LJ) interactions. Polarizability and long-range LJ interactions increase the membrane surface viscosity, which decreases the translational diffusion constants. Simulated diffusion constants for dipalmitoylphosphatidylcholine (DPPC) and dioleoylphosphatidylcholine (DOPC) extrapolated to infinite system size agree well with experiment for Drude2023, but overestimate experiment by 60% (on average) and a factor of 2.5 for C36/LJ-PME and C36, respectively. The relaxation time of lipid wobble is described about equally well by C36/LJ-PME and Drude2023, as consistent with the hexadecane viscosity for the FF, and both are more accurate than C36. Hence, physical improvements in the FF, which slowed down these dynamic processes led to better agreement with experiment. Lastly, bilayers containing 10 influenza fusion peptides and high fractions of lysolipids (which are known to accelerate pore formation) were simulated with C36 and Drude2023. Pore formation rates were comparable for the two FF for the bilayers with 80% and 90% lysolipid. However, while no pores formed in 24 μs (including a single 20 μs trajectory) in the 70% lysolipid with C36, 4 of 15 replicates formed pores in less than 1 μs with Drude. While the pathway to poration is qualitatively similar for the additive and polarizable FF for the systems studied, Drude2023 should be considered for quantitative studies of pore formation, and, in some cases, will accelerate the process.
Collapse
Affiliation(s)
- Richard M. Venable
- Laboratory of Computational Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Anthony J. Pane
- Laboratory of Computational Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Amy Rice
- Laboratory of Computational Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Richard W. Pastor
- Laboratory of Computational Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
182
|
Jiang Y, Wang Z, Scheuring S. A structural biology compatible file format for atomic force microscopy. Nat Commun 2025; 16:1671. [PMID: 39955301 PMCID: PMC11829953 DOI: 10.1038/s41467-025-56760-7] [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: 11/14/2024] [Accepted: 01/30/2025] [Indexed: 02/17/2025] Open
Abstract
Cryogenic electron microscopy (cryo-EM), X-ray crystallography, and nuclear magnetic resonance (NMR) contribute structural data that are interchangeable, cross-verifiable, and visualizable on common platforms, making them powerful tools for our understanding of protein structures. Unfortunately, atomic force microscopy (AFM) has so far failed to interface with these structural biology methods, despite the recent development of localization AFM (LAFM) that allows extracting high-resolution structural information from AFM data. Here, we build on LAFM and develop a pipeline that transforms AFM data into 3D-density files (.afm) that are readable by programs commonly used to visualize, analyze, and interpret structural data. We show that 3D-LAFM densities can serve as force fields to steer molecular dynamics flexible fitting (MDFF) to obtain structural models of previously unresolved states based on AFM observations in close-to-native environment. Besides, the .afm format enables direct 3D or 2D visualization and analysis of conventional AFM images. We anticipate that the file format will find wide usage and embed AFM in the repertoire of methods routinely used by the structural biology community, allowing AFM researchers to deposit data in repositories in a format that allows comparison and cross-verification with data from other techniques.
Collapse
Affiliation(s)
- Yining Jiang
- Biochemistry & Structural Biology, Cell & Developmental Biology, and Molecular Biology (BCMB) Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
- Weill Cornell Medicine, Department of Anesthesiology, New York, NY, USA
| | - Zhaokun Wang
- Weill Cornell Medicine, Department of Anesthesiology, New York, NY, USA
- Physiology, Biophysics and Systems Biology Graduate Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Simon Scheuring
- Weill Cornell Medicine, Department of Anesthesiology, New York, NY, USA.
- Weill Cornell Medicine, Department of Physiology and Biophysics, New York, NY, USA.
| |
Collapse
|
183
|
Pan D, Li R, Yang X, Yang X, Xu Z. Size dependence of the ion pairing preferences investigated by free energy calculations. J Chem Phys 2025; 162:064501. [PMID: 39927537 DOI: 10.1063/5.0248154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/22/2025] [Indexed: 02/11/2025] Open
Abstract
We calculate pairing free energies for a series of model monovalent ion pairs with equal sizes spanning a large radius range in the aqueous solution. Thermodynamic analysis reveals that the ion pairing strength displays an initial decrease in a nearly linear fashion followed by a significant enhancement at a relatively slower rate as the ion size gradually increases, resulting in the weakest pairing preference for ions with intermediate size. The free energy decomposition illustrates that the ion pairing is jointly governed by a delicate balance of the favorable ion-ion interaction term and the repulsive solvent-induced contribution, with their compensation leading to a small pairing free energy. The dependence of this substantial compensation on the ion size actually dominates the ion pairing strength and the magnitude of the free energy, which accounts for the occurrence of the less association between the intermediate-size ions. Estimation of van der Waals and electrostatic free energies highlights the importance of attractive dispersion forces in determining the shape of the potential of mean force, specifically for large ions with less favorable interaction with water molecules than the water-water interactions. In addition, the effect of varying the cation size on the decreased pairing preference surpasses that of altering the anion size. Our study significantly enhances our understanding of the empirical rule of matching ion size for predicting ion pairing preferences in aqueous solutions.
Collapse
Affiliation(s)
- Dan Pan
- College of Chemical Engineering, State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Rui Li
- College of Chemical Engineering, State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Xiao Yang
- College of Chemical Engineering, State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Xiaoning Yang
- College of Chemical Engineering, State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Zhijun Xu
- College of Chemical Engineering, State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China
- NJTECH University Suzhou Future Membrane Technology Innovation Center, Suzhou 215300, China
| |
Collapse
|
184
|
Subramaniam T, Mualif SA, Chan WH, Abd Halim KB. Unlocking the potential of in silico approach in designing antibodies against SARS-CoV-2. FRONTIERS IN BIOINFORMATICS 2025; 5:1533983. [PMID: 40017562 PMCID: PMC11865036 DOI: 10.3389/fbinf.2025.1533983] [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: 11/25/2024] [Accepted: 01/17/2025] [Indexed: 03/01/2025] Open
Abstract
Antibodies are naturally produced safeguarding proteins that the immune system generates to fight against invasive invaders. For centuries, they have been produced artificially and utilized to eradicate various infectious diseases. Given the ongoing threat posed by COVID-19 pandemics worldwide, antibodies have become one of the most promising treatments to prevent infection and save millions of lives. Currently, in silico techniques provide an innovative approach for developing antibodies, which significantly impacts the formulation of antibodies. These techniques develop antibodies with great specificity and potency against diseases such as SARS-CoV-2 by using computational tools and algorithms. Conventional methods for designing and developing antibodies are frequently costly and time-consuming. However, in silico approach offers a contemporary, effective, and economical paradigm for creating next-generation antibodies, especially in accordance with recent developments in bioinformatics. By utilizing multiple antibody databases and high-throughput approaches, a unique antibody construct can be designed in silico, facilitating accurate, reliable, and secure antibody development for human use. Compared to their traditionally developed equivalents, a large number of in silico-designed antibodies have advanced swiftly to clinical trials and became accessible sooner. This article helps researchers develop SARS-CoV-2 antibodies more quickly and affordably by giving them access to current information on computational approaches for antibody creation.
Collapse
Affiliation(s)
- Tasshitra Subramaniam
- Biomedical Engineering and Health Sciences Department, Faculty of Electrical Engineering, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
| | - Siti Aisyah Mualif
- Biomedical Engineering and Health Sciences Department, Faculty of Electrical Engineering, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
- Advanced Diagnostics and Progressive Human Care, Biomedical Engineering and Health Sciences Department, Faculty of Electrical Engineering, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
| | - Weng Howe Chan
- Faculty of Computing, Universiti Teknologi Malaysia, Johor Bahru, Johor, Malaysia
| | - Khairul Bariyyah Abd Halim
- Department of Biotechnology, Kulliyyah of Science, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
- Research Unit for Bioinformatics and Computational Biology (RUBIC), Kulliyyah of Science, International Islamic University Malaysia, Kuantan, Pahang, Malaysia
| |
Collapse
|
185
|
Hu Z, Martí J. Atomic-level mechanisms of abnormal activation in NRAS oncogenes from two-dimensional free energy landscapes. NANOSCALE 2025; 17:4047-4057. [PMID: 39775302 DOI: 10.1039/d4nr03372h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
The NRAS-mutant subset of melanoma is one of the most aggressive and lethal types associated with poor overall survival. Unfortunately, a low understanding of the NRAS-mutant dynamic behavior has led to the lack of clinically approved therapeutic agents able to directly target NRAS oncogenes. In this work, accurate local structures of NRAS and its mutants have been fully explored through the corresponding free energy surfaces obtained by microsecond scale well-tempered metadynamics simulations. Free energy calculations are crucial to reveal the precise mechanisms of Q61 mutations at the atomic level. Considering specific atom-atom distances d and angles ϕ as appropriate reaction coordinates we have obtained free energy surfaces revealing local and global minima together with their main transition states, unveiling the mechanisms of abnormal NRAS activation from the atomic-level and quantitatively analyzing the corresponding stable states. This will help in advancing our understanding of the basic mechanisms of NRAS mutations, offering new opportunities for the design of potential inhibitors.
Collapse
Affiliation(s)
- Zheyao Hu
- Department of Physics, Polytechnic University of Catalonia-Barcelona Tech, B5-209 Northern Campus, Jordi Girona 1-3, 08034 Barcelona, Catalonia, Spain.
| | - Jordi Martí
- Department of Physics, Polytechnic University of Catalonia-Barcelona Tech, B5-209 Northern Campus, Jordi Girona 1-3, 08034 Barcelona, Catalonia, Spain.
| |
Collapse
|
186
|
Clayton J, Romany A, Matenoglou E, Gavathiotis E, Poulikakos PI, Shen J. Mechanism of dimer selectivity and binding cooperativity of BRAF inhibitors. eLife 2025; 13:RP95334. [PMID: 39945510 PMCID: PMC11825127 DOI: 10.7554/elife.95334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2025] Open
Abstract
Aberrant signaling of BRAFV600E is a major cancer driver. Current FDA-approved RAF inhibitors selectively inhibit the monomeric BRAFV600E and suffer from tumor resistance. Recently, dimer-selective and equipotent RAF inhibitors have been developed; however, the mechanism of dimer selectivity is poorly understood. Here, we report extensive molecular dynamics (MD) simulations of the monomeric and dimeric BRAFV600E in the apo form or in complex with one or two dimer-selective (PHI1) or equipotent (LY3009120) inhibitor(s). The simulations uncovered the unprecedented details of the remarkable allostery in BRAFV600E dimerization and inhibitor binding. Specifically, dimerization retrains and shifts the αC helix inward and increases the flexibility of the DFG motif; dimer compatibility is due to the promotion of the αC-in conformation, which is stabilized by a hydrogen bond formation between the inhibitor and the αC Glu501. A more stable hydrogen bond further restrains and shifts the αC helix inward, which incurs a larger entropic penalty that disfavors monomer binding. This mechanism led us to propose an empirical way based on the co-crystal structure to assess the dimer selectivity of a BRAFV600E inhibitor. Simulations also revealed that the positive cooperativity of PHI1 is due to its ability to preorganize the αC and DFG conformation in the opposite protomer, priming it for binding the second inhibitor. The atomically detailed view of the interplay between BRAF dimerization and inhibitor allostery as well as cooperativity has implications for understanding kinase signaling and contributes to the design of protomer selective RAF inhibitors.
Collapse
Affiliation(s)
- Joseph Clayton
- Department of Pharmaceutical Sciences, University of Maryland School of PharmacyBaltimoreUnited States
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, United States Food and Drug AdministrationSilver SpringUnited States
| | - Aarion Romany
- Department of Pharmaceutical Sciences, University of Maryland School of PharmacyBaltimoreUnited States
| | - Evangelia Matenoglou
- Department of Biochemistry, Department of Medicine, Department of Oncology, Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of MedicineNew YorkUnited States
| | - Evripidis Gavathiotis
- Department of Biochemistry, Department of Medicine, Department of Oncology, Montefiore Einstein Comprehensive Cancer Center, Albert Einstein College of MedicineNew YorkUnited States
| | - Poulikos I Poulikakos
- Department of Oncological Sciences, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Jana Shen
- Department of Pharmaceutical Sciences, University of Maryland School of PharmacyBaltimoreUnited States
| |
Collapse
|
187
|
Ektirici S, Harmandaris V. A study of alpha-synuclein and poly( N-isopropylacrylamide) complex formation through detailed atomistic simulations. SOFT MATTER 2025; 21:1382-1394. [PMID: 39868549 DOI: 10.1039/d4sm01395f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
This work presents an investigation of the influence of poly(N-isopropylacrylamide) (PNIPAM) polymer on the structural dynamics of intrinsically disordered alpha-synuclein (α-syn) protein, exploring the formation and intricate features of the resulting α-syn/PNIPAM complexes. Using atomistic molecular dynamics (MD) simulations, our study analyzes the impact of initial configuration, polymer molecular weight, and protein mutations on the α-syn and the α-syn/PNIPAM complex. Atomistic simulations, of a few μs, of the protein/polymer complex reveal crucial insights into molecular interactions within the complex, emphasizing a delicate balance of forces governing its stability and structural evolution. Our findings indicate that PNIPAM polymer engages in significant non-polar interactions with the non-amyloid component (NAC) region of α-syn, which plays a crucial role in fibril formation, under various conditions such as the mutations in the protein structure and polymer chain length. Especially the PNIPAM polymer with a 40mer monomer exhibits a stabilizing effect on the structural properties of the protein, reducing intramolecular interactions that contribute to misfolding. These findings, which delve into protein/polymer interactions, hold promise as potential guidance for therapeutic strategies in various neurodegenerative disorders.
Collapse
Affiliation(s)
- Sisem Ektirici
- Computation-based Science and Technology Research Center, The Cyprus Institute, 2121 Nicosia, Cyprus.
| | - Vagelis Harmandaris
- Computation-based Science and Technology Research Center, The Cyprus Institute, 2121 Nicosia, Cyprus.
- Department of Mathematics and Applied Mathematics, University of Crete, Heraklion, GR-71110, Greece
- Institute of Applied and Computational Mathematics, Foundation for Research and Technology - Hellas, Heraklion, GR-71110 Crete, Greece
| |
Collapse
|
188
|
Thompson RD, Carbaugh DL, Nielsen JR, Witt CM, Faison EM, Meganck RM, Rangadurai A, Zhao B, Bonin JP, Nicely NI, Marzluff WF, Frank AT, Lazear HM, Zhang Q. Lifetime of ground conformational state determines the activity of structured RNA. Nat Chem Biol 2025:10.1038/s41589-025-01843-1. [PMID: 39939412 DOI: 10.1038/s41589-025-01843-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 01/15/2025] [Indexed: 02/14/2025]
Abstract
Biomolecules continually sample alternative conformations. Consequently, even the most energetically favored ground conformational state has a finite lifetime. Here, we show that, in addition to the three-dimensional (3D) structure, the lifetime of a ground conformational state determines its biological activity. Using hydrogen-deuterium exchange nuclear magnetic resonance spectroscopy, we found that Zika virus exoribonuclease-resistant RNA (xrRNA) encodes a ground conformational state with a lifetime that is ~105-107 longer than that of canonical base pairs. Mutations that shorten the apparent lifetime of the ground state without affecting its 3D structure decreased exoribonuclease resistance in vitro and impaired virus replication in cells. Additionally, we observed this exceptionally long-lived ground state in xrRNAs from diverse infectious mosquito-borne flaviviruses. These results demonstrate the biological importance of the lifetime of a preorganized ground state and further suggest that elucidating the lifetimes of dominant 3D structures of biomolecules may be crucial for understanding their behaviors and functions.
Collapse
Affiliation(s)
- Rhese D Thompson
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Derek L Carbaugh
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joshua R Nielsen
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ciara M Witt
- Department of Biophysics and Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Edgar M Faison
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rita M Meganck
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Atul Rangadurai
- Department of Biochemistry, Duke University, Durham, NC, USA
- NanoVation Therapeutics, Vancouver, British Columbia, Canada
| | - Bo Zhao
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jeffrey P Bonin
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nathan I Nicely
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - William F Marzluff
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Aaron T Frank
- Department of Biophysics and Chemistry, University of Michigan, Ann Arbor, MI, USA.
- Arrakis Therapeutics, Waltham, MA, USA.
| | - Helen M Lazear
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Qi Zhang
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- RNA Discovery Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
189
|
Niu Z, Kementzidis G, Rafailovich M, Simon M, Papadopoulos E, Aktas BH, Deng Y. Simulations of pH and thermal effects on SARS-CoV-2 spike glycoprotein. Front Mol Biosci 2025; 12:1545041. [PMID: 40007557 PMCID: PMC11850259 DOI: 10.3389/fmolb.2025.1545041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
We performed triplicate and long-time all-atom molecular dynamics simulations to investigate the structures and dynamics of the SARS-CoV-2 spike glycoprotein (S-protein) for a broad range of pH = 1 through 11 and temperatures of 3°C through 75°C. This study elucidates the complex interplay between pH and thermal effects on S-protein structures, with implications for its behavior under diverse conditions, and identifies the RBD as a primary region of the structural deviations. We found: 1) Structural deviations in the S-protein backbone at pH = 1 are 210% greater than those at pH = 7 at 75°C, with most of the deviations appearing in the receptor-binding domain (RBD). Smaller structural changes are observed at pH = 3 and 11. 2) The pH and thermal conditions impact on the protein structures: substantial acidic and basic conditions expand the protein's solvent exposure, while high heat contracts. This effect is primarily pH-driven at extreme acidity and thermo-driven at moderate pH. 3) The Gibbs free energy landscape reveals that pH as the main driver of structural changes. 4) The parametrized methods enable the predictions of the S-protein properties at any reasonable pH and thermal conditions without explicit MD simulations.
Collapse
Affiliation(s)
- Ziyuan Niu
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, United States
| | - Georgios Kementzidis
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, United States
| | - Miriam Rafailovich
- Department of Materials Science and Chemical Engineering, Stony Brook University, Stony Brook, NY, United States
| | - Marcia Simon
- Department of Oral Biology and Pathology, Stony Brook University, Stony Brook, NY, United States
| | - Evangelos Papadopoulos
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Bertal H. Aktas
- Division of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Yuefan Deng
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
190
|
Möller D, van der Walt M, Oosthuizen C, Serian M, Serem JC, Lorenz CD, Mason AJ, Bester MJ, Gaspar ARM. Improving the Activity and Selectivity of a Scorpion-Derived Peptide, A3a, against Acinetobacter baumannii through Rational Design. ACS OMEGA 2025; 10:4699-4710. [PMID: 39959037 PMCID: PMC11822712 DOI: 10.1021/acsomega.4c09593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 02/18/2025]
Abstract
The rise in antimicrobial resistance has led to an increased desire to understand how antimicrobial peptides (AMPs) can be better engineered to kill antibiotic-resistant bacteria. Previously, we showed that C-terminal amidation of a peptide, identified in scorpion Androctonus amoreuxi venom, increased its activity against both Gram-positive and -negative bacteria. Here, we incorporate all-atom molecular dynamics (MD) simulations in a rational design strategy to create analogues of A3a with greater therapeutic potential. We discover two novel AMPs which achieve greater potency against, and selectivity toward, Acinetobacter baumannii ATCC 19606 but via two distinct mechanisms and which are effective in Galleria mellonella models of A. baumannii burn wound infection. While CD spectroscopy indicates A3a adopts an α-helix conformation in the presence of models of the Gram-negative bacterial plasma membrane, MD simulations reveal it adopts a hairpin conformation during initial binding. Three different strategies, designed to stabilize this hairpin conformation, produce substantially different outcomes. Deletion of Ile6 and Ile10 restricts conformational flexibility, characteristic of A3a, during membrane binding, prevents adoption of the α-helix conformation in the steady state, and abrogates the antibacterial activity. In contrast, substitution of arginine 7 to lysine (A3a[R7K]) or isoleucine 14 to tryptophan (A3a[I14W]) does not consistently affect peptide conformations. Both of these new analogues are rapidly bactericidal toward A. baumannii ATCC 19606 but A3a[R7K] also causes rapid permeabilization and while the antibacterial potency and selectivity are increased for both peptides, this is greatest for A3a[I14W]. Integration of atomistic MD simulations into a multidisciplinary approach to understanding antimicrobial peptide mechanism of action is a valuable tool for interpreting the effects of rational design strategies.
Collapse
Affiliation(s)
- Dalton
S. Möller
- Department
of Biochemistry, Genetics and Microbiology, Faculty of Natural and
Agricultural Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Mandelie van der Walt
- Department
of Biochemistry, Genetics and Microbiology, Faculty of Natural and
Agricultural Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Carel Oosthuizen
- Drug
Discovery and Development Centre (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Miruna Serian
- Department
of Physics, Faculty of Natural, Mathematical and Engineering Sciences, King’s College London, London WC2R 2LS, U.K.
| | - June C. Serem
- Department
of Anatomy, Faculty of Health Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Christian D. Lorenz
- Department
of Engineering, Faculty of Natural, Mathematical and Engineering Sciences, King’s College London, London WC2R 2LS, U.K.
| | - A. James Mason
- Institute
of Pharmaceutical Science, School of Cancer & Pharmaceutical Science,
Faculty of Life Sciences & Medicine, King’s College London, London SE1 9NH, U.K.
| | - Megan J. Bester
- Department
of Anatomy, Faculty of Health Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Anabella R. M. Gaspar
- Department
of Biochemistry, Genetics and Microbiology, Faculty of Natural and
Agricultural Sciences, University of Pretoria, Pretoria 0002, South Africa
| |
Collapse
|
191
|
Rivera-Suárez BA, García González VG, Chimal-Vega B, Navarro Padrón AC, Galindo-Hernández O, Vique-Sánchez JL. Potential Ligands to Resistin Against Prostate Cancer, Evaluated by Molecular Docking and In Vitro Assays to Develop an Anticancer Drug. Chem Biodivers 2025:e202500189. [PMID: 39932405 DOI: 10.1002/cbdv.202500189] [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: 01/15/2025] [Revised: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 02/19/2025]
Abstract
Overweight and obesity are risk factors that increase the white adipose tissue (WAT) that promotes the release of adipokines (adiponectin, leptin, and resistin). The increase of the resistin levels contributes to different cellular processes; regulation in the metabolism, inflammation process, and particularly in messengers in some cancer types. Resistin promotes cell proliferation and migration. Therefore, resistin is proposed as a multipotential therapeutic target to treat different diseases, and in this study, we focus on the regulation of resistin and its effect on prostate cancer. This study proposes compounds selective to resistin, these were selected and evaluated by molecular docking and in vitro assays, to develop a new drug against the resistin´s functions related to interaction with their potential receptors (Δ-DCN, TLR4, and CAP-1). These molecules with pharmacological characteristics are capable of interacting in the regions of resistin to hinder/block the interaction between resistin and their possible receptors (Δ-DCN, TLR4, and CAP-1). We determined two compounds that showed to be selective against resistin by in vitro assays. In this way, this study proposes compounds that were developed to be selective against resistin, and it could decrease the effect known of resistin by their receptors related to the proliferation of cancer.
Collapse
Affiliation(s)
- Bryan A Rivera-Suárez
- Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali, Mexico
| | | | - Brenda Chimal-Vega
- Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali, Mexico
| | - Anna C Navarro Padrón
- Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali, Mexico
| | | | - José L Vique-Sánchez
- Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali, Mexico
| |
Collapse
|
192
|
Mukhaleva E, Manookian B, Chen H, Sivaraj IR, Ma N, Wei W, Urbaniak K, Gogoshin G, Bhattacharya S, Vaidehi N, Rodin AS, Branciamore S. BaNDyT: Bayesian Network Modeling of Molecular Dynamics Trajectories. J Chem Inf Model 2025; 65:1278-1288. [PMID: 39846243 DOI: 10.1021/acs.jcim.4c01981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Bayesian network modeling (BN modeling, or BNM) is an interpretable machine learning method for constructing probabilistic graphical models from the data. In recent years, it has been extensively applied to diverse types of biomedical data sets. Concurrently, our ability to perform long-time scale molecular dynamics (MD) simulations on proteins and other materials has increased exponentially. However, the analysis of MD simulation trajectories has not been data-driven but rather dependent on the user's prior knowledge of the systems, thus limiting the scope and utility of the MD simulations. Recently, we pioneered using BNM for analyzing the MD trajectories of protein complexes. The resulting BN models yield novel fully data-driven insights into the functional importance of the amino acid residues that modulate proteins' function. In this report, we describe the BaNDyT software package that implements the BNM specifically attuned to the MD simulation trajectories data. We believe that BaNDyT is the first software package to include specialized and advanced features for analyzing MD simulation trajectories using a probabilistic graphical network model. We describe here the software's uses, the methods associated with it, and a comprehensive Python interface to the underlying generalist BNM code. This provides a powerful and versatile mechanism for users to control the workflow. As an application example, we have utilized this methodology and associated software to study how membrane proteins, specifically the G protein-coupled receptors, selectively couple to G proteins. The software can be used for analyzing MD trajectories of any protein as well as polymeric materials.
Collapse
Affiliation(s)
- Elizaveta Mukhaleva
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, 1218 S 5th Ave, Monrovia, California 91016, United States
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, California 91010, United States
| | - Babgen Manookian
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, 1218 S 5th Ave, Monrovia, California 91016, United States
| | - Hanyu Chen
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, 1218 S 5th Ave, Monrovia, California 91016, United States
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, California 91010, United States
| | - Indira R Sivaraj
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, 1218 S 5th Ave, Monrovia, California 91016, United States
| | - Ning Ma
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, 1218 S 5th Ave, Monrovia, California 91016, United States
| | - Wenyuan Wei
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, 1218 S 5th Ave, Monrovia, California 91016, United States
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, California 91010, United States
| | - Konstancja Urbaniak
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, 1218 S 5th Ave, Monrovia, California 91016, United States
| | - Grigoriy Gogoshin
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, 1218 S 5th Ave, Monrovia, California 91016, United States
| | - Supriyo Bhattacharya
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, 1218 S 5th Ave, Monrovia, California 91016, United States
| | - Nagarajan Vaidehi
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, 1218 S 5th Ave, Monrovia, California 91016, United States
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, California 91010, United States
| | - Andrei S Rodin
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, 1218 S 5th Ave, Monrovia, California 91016, United States
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, California 91010, United States
| | - Sergio Branciamore
- Department of Computational and Quantitative Medicine, Beckman Research Institute of the City of Hope, 1218 S 5th Ave, Monrovia, California 91016, United States
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of the City of Hope, 1500 E Duarte Road, Duarte, California 91010, United States
| |
Collapse
|
193
|
Catalina-Hernandez E, Aguilella-Arzo M, Peralvarez-Marin A, Lopez-Martin M. Computational Insights into Membrane Disruption by Cell-Penetrating Peptides. J Chem Inf Model 2025; 65:1549-1559. [PMID: 39823544 PMCID: PMC11815844 DOI: 10.1021/acs.jcim.4c01940] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/15/2024] [Accepted: 12/30/2024] [Indexed: 01/19/2025]
Abstract
Cell-penetrating peptides (CPPs) can translocate into cells without inducing cytotoxicity. The internalization process implies several steps at different time scales ranging from microseconds to minutes. We combine adaptive Steered Molecular Dynamics (aSMD) with conventional Molecular Dynamics (cMD) to observe nonequilibrium and equilibrium states to study the early mechanisms of peptide-bilayer interaction leading to CPPs internalization. We define three membrane compositions representing bilayer sections, neutral lipids (i.e., upper leaflet), neutral lipids with cholesterol (i.e., hydrophobic core), and neutral/negatively charged lipids with cholesterol (i.e., lower leaflet) to study the energy barriers and disruption mechanisms of Arg9, MAP, and TP2, representing cationic, amphiphilic, and hydrophobic CPPs, respectively. Cholesterol and negatively charged lipids increase the energetic barriers for the peptide-bilayer crossing. TP2 interacts with the bilayer by hydrophobic insertion, while Arg9 disrupts the bilayer by forming transient or stable pores. MAP has shown both behaviors. Collectively, these findings underscore the significance of innovative computational approaches in studying membrane-disruptive peptides and, more specifically, in harnessing their potential for cell penetration.
Collapse
Affiliation(s)
- Eric Catalina-Hernandez
- Unit
of Biophysics, Department of Biochemistry and Molecular Biology, Facultat
de Medicina, Av. Can Domènech s/n, Universitat Autònoma de Barcelona, 08193 Cerdanyola del
Vallès, Catalonia, Spain
- Institute
of Neurosciences, Universitat Autònoma
de Barcelona, 08193 Cerdanyola del Vallès, Catalonia, Spain
| | - Marcel Aguilella-Arzo
- Laboratory
of Molecular Biophysics, Department of Physics, University Jaume I, 12071 Castellon, Spain
| | - Alex Peralvarez-Marin
- Unit
of Biophysics, Department of Biochemistry and Molecular Biology, Facultat
de Medicina, Av. Can Domènech s/n, Universitat Autònoma de Barcelona, 08193 Cerdanyola del
Vallès, Catalonia, Spain
- Institute
of Neurosciences, Universitat Autònoma
de Barcelona, 08193 Cerdanyola del Vallès, Catalonia, Spain
| | - Mario Lopez-Martin
- Unit
of Biophysics, Department of Biochemistry and Molecular Biology, Facultat
de Medicina, Av. Can Domènech s/n, Universitat Autònoma de Barcelona, 08193 Cerdanyola del
Vallès, Catalonia, Spain
- Institute
of Neurosciences, Universitat Autònoma
de Barcelona, 08193 Cerdanyola del Vallès, Catalonia, Spain
| |
Collapse
|
194
|
Chen L, Leung JMG, Zsigmond K, Chong LT, Miranda-Quintana RA. SHINE: Deterministic Many-to-Many clustering of Molecular Pathways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.636541. [PMID: 39975301 PMCID: PMC11839051 DOI: 10.1101/2025.02.07.636541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
State-of-the-art molecular dynamics (MD) simulation methods can generate diverse ensembles of pathways for complex biological processes. Analyzing these pathways using statistical mechanics tools demands identifying key states that contribute to both the dynamic and equilibrium properties of the system. This task becomes especially challenging when analyzing multiple MD simulations simultaneously, a common scenario in enhanced sampling techniques like the weighted ensemble strategy. Here, we present a new module of the MDANCE package designed to streamline the analysis of pathway ensembles. This module integrates n-ary similarity, cheminformatics-inspired tools, and hierarchical clustering to improve analysis efficiency. We present the theoretical foundation behind this approach, termed Sampling Hierarchical Intrinsic N-ary Ensembles (SHINE), and demonstrate its application to simulations of alanine dipeptide and adenylate kinase.
Collapse
Affiliation(s)
- Lexin Chen
- Department of Chemistry and Quantum Theory Project, University of Florida, Gainesville, FL 32603, USA
| | - Jeremy M G Leung
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Krisztina Zsigmond
- Department of Chemistry and Quantum Theory Project, University of Florida, Gainesville, FL 32603, USA
| | - Lillian T Chong
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | | |
Collapse
|
195
|
Nahian M, Khan MR, Rahman F, Reza HM, Bayil I, Nodee TA, Basher T, Sany MR, Munmun RN, Habib SMA, Mazumder L, Acharjee M. Immunoinformatic strategy for developing multi-epitope subunit vaccine against Helicobacter pylori. PLoS One 2025; 20:e0318750. [PMID: 39919064 PMCID: PMC11805379 DOI: 10.1371/journal.pone.0318750] [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: 08/11/2024] [Accepted: 01/20/2025] [Indexed: 02/09/2025] Open
Abstract
Helicobacter pylori is a gram-negative bacterium that persistently infects the human stomach, leading to peptic ulcers, gastritis, and an increased risk of gastric cancer. The extremophilic characteristics of this bacterium make it resistant to current drug treatments, and there are no licensed vaccines available against H. pylori. Computational approaches offer a viable alternative for designing antigenic, stable, and safe vaccines to control infections caused by this pathogen. In this study, we employed an immunoinformatic strategy to design a set of candidate multi-epitope subunit vaccines by combining the most potent B and T cell epitopes from three targeted antigenic proteins (BabA, CagA, and VacA). Out of the 12 hypothetical vaccines generated, two (HP_VaX_V1 and HP_VaX_V2) were found to be strongly immunogenic, non-allergenic, and structurally stable. The proposed vaccine candidates were evaluated based on population coverage, molecular docking, immune simulations, codon adaptation, secondary mRNA structure, and in silico cloning. The vaccine candidates exhibited antigenic scores of 1.19 and 1.01, with 93.5% and 90.4% of the most rama-favored regions, respectively. HP_VaX_V1 and HP_VaX_V2 exhibited the strongest binding affinity towards TLR-7 and TLR-8, as determined by molecular docking simulations (ΔG = -20.3 and -20.9, respectively). Afterward, multi-scale normal mode analysis simulation revealed the structural flexibility and stability of vaccine candidates. Additionally, immune simulations showed elevated levels of cell-mediated immunity, while repeated exposure simulations indicated rapid antigen clearance. Finally, in silico cloning was performed using the expression vector pET28a (+) with optimized restriction sites to develop a viable strategy for large-scale production of the chosen vaccine constructs. These analyses suggest that the proposed vaccines may elicit potent immune responses against H. pylori, but laboratory validation is needed to verify their safety and immunogenicity.
Collapse
Affiliation(s)
- Md. Nahian
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | - Md. Rasel Khan
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | - Fabiha Rahman
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | - Hossain Mohammed Reza
- Faculty of Life and Health Sciences, School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, Northern Ireland
| | - Imren Bayil
- Department of Bioinformatics and Computational Biology, Gaziantep University, Gaziantep, Turkey
| | | | - Tabassum Basher
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | | | | | | | - Lincon Mazumder
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | - Mrityunjoy Acharjee
- Department of Microbiology, Stamford University Bangladesh, Dhaka, Bangladesh
| |
Collapse
|
196
|
Xu Z, Gan M, Guan W, Tian F, Wang Y, Zhang J, Cai L. In Slico Screening and In Vitro Identification of Hyperuricemia-Inhibiting Peptides from Trachurus japonicus. Foods 2025; 14:524. [PMID: 39942117 PMCID: PMC11817512 DOI: 10.3390/foods14030524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Hyperuricemia arises from imbalanced uric acid metabolism, contributing to gout and related chronic diseases. When traditional drugs are used to treat hyperuricemia, side effects are inevitable, which promotes the exploration of new bioactive compounds. Protein hydrolysates and peptides are gradually showing potential in the treatment of hyperuricemia. This study investigated the uric acid inhibitory activity of peptides extracted from Trachurus japonicus using in silico and in vitro methods. We employed in silico virtual enzymolysis and experimental validation to identify bioactive peptides from Trachurus japonicus proteins. Four peptides (DF, AGF, QPSF, and AGDDAPR) were comprehensively screened by molecular docking and database analysis. After solid-phase synthesis, the inhibitory effects of these peptides on hyperuricemia were further verified in vitro and at the cellular level. The results showed that all four peptides have good hyperuricemia-inhibiting activities. Molecular docking and molecular dynamics revealed that peptides DF and AGDDAPR affect the production of uric acid by binding to the active sites of urate transporter 1 (URAT1), glucose transporter 9 (GLUT9), and xanthine oxidase (XOD), while peptides QPSF and AGF mainly influence the XOD active site, confirming that it is feasible to rapidly screen hyperuricemia-inhibiting peptides by molecular docking.
Collapse
Affiliation(s)
- Zexuan Xu
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, China;
| | - Miaoyu Gan
- Ningbo Innovation Center, College of Biosystems and Food Science, Zhejiang University, Ningbo 315100, China; (M.G.); (Y.W.)
| | - Weiliang Guan
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China;
| | - Fang Tian
- Key Laboratory of Health Risk Factors for Seafood of Zhejiang Province, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China;
| | - Yuxi Wang
- Ningbo Innovation Center, College of Biosystems and Food Science, Zhejiang University, Ningbo 315100, China; (M.G.); (Y.W.)
| | - Jinjie Zhang
- College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo 315211, China;
| | - Luyun Cai
- Ningbo Innovation Center, College of Biosystems and Food Science, Zhejiang University, Ningbo 315100, China; (M.G.); (Y.W.)
| |
Collapse
|
197
|
Momin Y, Beloshe V. Pharmacophore modeling in drug design. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2025; 103:313-324. [PMID: 40175047 DOI: 10.1016/bs.apha.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
A successful and expanded area of computational drug design is pharmacophore modeling. A pharmacophore is a description of the structural features of a compound that are essential to its biological activity. The rational design of new drugs has made extensive use of the pharmacophore concept. By schematically illustrating the essential components of molecular recognition, Pharmacophores can be used to represent and identify molecules in two or three dimensions. Besides target identification, the pharmacophore concept is also helpful for side effects, off-target, and absorption, distribution, and toxicity modeling. Moreover, to enhance virtual screening, pharmacophores, and molecular docking simulations are frequently coupled. A completely new area of drug design has been made possible by the development of machine learning techniques and pharmacophore mapping algorithms, wherein an ineffective molecule with the right modifications may have the potential to function as an inhibitor. This approach has been stimulated by its predictive abilities to assess the possibility that a set of compounds will be active against protein targets of interest. With alignment to the standard pharmacophore model, active compounds of the protein target can be developed. The pharmacophore modeling/screening technique is used to identify possible proteins of interest and seek out/suggest novel therapeutic uses for the drug.
Collapse
Affiliation(s)
- Yasmin Momin
- Department of Pharmaceutical Chemistry, Annasaheb Dange College of B. Pharmacy, Ashta, Maharashtra, India.
| | | |
Collapse
|
198
|
Avery N, Childers KC, McCarty J, Spiegel PC. Atomistic Mechanism of Lipid Membrane Binding for Blood Coagulation Factor VIII with Molecular Dynamics Simulations on a Microsecond Time Scale. J Phys Chem B 2025; 129:1486-1498. [PMID: 39840640 PMCID: PMC11808648 DOI: 10.1021/acs.jpcb.4c06575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/04/2025] [Accepted: 01/10/2025] [Indexed: 01/23/2025]
Abstract
During the blood coagulation cascade, coagulation factor VIII (FVIII) is activated by thrombin to form activated factor VIII (FVIIIa). FVIIIa associates with platelet surfaces at the site of vascular damage to form an intrinsic tenase complex with activated factor IX. A working model for FVIII membrane binding involves the association of positively charged FVIII residues with negatively charged lipid headgroups and the burial of hydrophobic residues into the membrane interior. Currently, the atomic details of the FVIII lipid binding interactions and membrane orientation are lacking. This study reports residue-specific FVIII C domain interactions with 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) and 1,2-dioleoyl-sn-glycero-3-phospho-l-serine (DOPS) in atomistic detail. Contact maps between residues in the C domains with different lipid moieties support prior structural data describing how the C domains associate with membranes through electrostatic and hydrophobic interactions. Solvent-accessible surface area analysis quantified the extent to which residues in the C1 and C2 domains bury into the membrane. Calculations of the potential energy between the C domains and DOPC and DOPS revealed an FVIII membrane-binding orientation that agrees with previous experimental data. This study expands our knowledge of the structural basis of FVIII membrane association, which may be critical for the development of next-generation FVIII replacement constructs with improved activity.
Collapse
Affiliation(s)
- Nathan
G. Avery
- Chemistry Department, Western Washington University, Bellingham, Washington 98225-9038, United
States
| | | | - James McCarty
- Chemistry Department, Western Washington University, Bellingham, Washington 98225-9038, United
States
| | - Paul Clinton Spiegel
- Chemistry Department, Western Washington University, Bellingham, Washington 98225-9038, United
States
| |
Collapse
|
199
|
Cheon SY, Kim YE, Yang ES, Lim YJ, Bae CH, Jin JS, Park W, Kim BS, Kim C, Cho H, Kim S, Lee SH, Ha KT. Synthesis of 1-Hydroxy(and 1-Alkoxy, 1-Acyloxy)-1H-indoles and evaluations of their suppressive activities against tumor growth through inhibiting lactate dehydrogenase A. Eur J Med Chem 2025; 283:117104. [PMID: 39642694 DOI: 10.1016/j.ejmech.2024.117104] [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: 10/07/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 12/09/2024]
Abstract
Inhibition of lactate dehydrogenase (LDH) has emerged as a promising cancer therapy strategy due to its essential role in the metabolic transformation of cancer cells. In this study, 53 derivatives of 1-hydroxy(and 1-alkoxy, 1-acyloxy)indoles were designed, synthesized, and biologically evaluated. Several multi-substituted 1-hydroxy(and 1-alkoxy, 1-acyloxy)indole compounds exhibited inhibitory activity against the LDH-A isoform (LDHA). We confirmed that the C(3)-substituent provided additional significant hydrogen bonding and hydrophobic interactions, which enhanced the LDHA inhibitory activity with high selectivity. Our results revealed that methyl 4-bromo-3-[(n-hexyloxy)methyl]-1-hydroxy-1H-indole-2-carboxylate (1g), selectively inhibited LDHA (IC50 = 25 ± 1.12 nM) without affecting the LDH-B isoform (LDHB). The compound exhibited potent cytotoxic activity in several cancer cell lines, including DLD-1 colorectal cancer cells (GI50 = 27 ± 1.2 μM). Compound 1g significantly inhibited cancer cell growth by activating apoptotic pathways in a xenograft cancer model, without causing weight loss or liver and kidney damage. Therefore, compound 1g may serve as a highly specific and promising candidate for the development of LDHA inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Se-Yun Cheon
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
| | - Ye Eun Kim
- College of Pharmacy and Innovative Drug Center, Duksung Women's University, Seoul, 01369, Republic of Korea
| | - Eun-Sun Yang
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
| | - Yoo Jin Lim
- College of Pharmacy and Innovative Drug Center, Duksung Women's University, Seoul, 01369, Republic of Korea
| | - Chang-Hwan Bae
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, Jeollanam-do, 58245, Republic of Korea
| | - Jung-Sook Jin
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
| | - Wonyoung Park
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea; Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
| | - Bo-Sung Kim
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea; Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
| | - Chorong Kim
- College of Pharmacy and Innovative Drug Center, Duksung Women's University, Seoul, 01369, Republic of Korea
| | - Hyunsung Cho
- College of Pharmacy and Innovative Drug Center, Duksung Women's University, Seoul, 01369, Republic of Korea
| | - Seungtae Kim
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea; Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea
| | - Sang Hyup Lee
- College of Pharmacy and Innovative Drug Center, Duksung Women's University, Seoul, 01369, Republic of Korea.
| | - Ki-Tae Ha
- Korean Medical Research Center for Healthy Aging, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea; Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongnam, 50612, Republic of Korea.
| |
Collapse
|
200
|
Feng R, Rafiei M, Fernando KS, Chau Y. Direct cytosol delivery of mRNA by micron-sized co-assembly with designer oligopeptides. J Mater Chem B 2025; 13:2167-2179. [PMID: 39803759 DOI: 10.1039/d4tb01098a] [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: 02/06/2025]
Abstract
Inefficient endosomal escape has been regarded as the main bottleneck for intracellular nucleic acid delivery. While most research efforts have been spent on designing various nano-sized particles, we took a different path here, investigating micron-sized carriers for direct cytosol entry. Using the spontaneous co-assembly of mRNA and the designer 27 amino acid oligopeptide named pepMAX2, micron-sized co-assemblies were obtained with various sizes by altering the concentration of NaCl salt and time for pre-incubation. Surprisingly, transfection was much more effective using micron-sized than nano-sized co-assemblies, and the efficiency surpasses that of a widely used lipid-based commercial reagent. The study was complemented by computational simulations, inhibitor studies and live-cell confocal imaging to shed light on the role of electrostatic interaction on assembly and the mechanism of uptake and intracellular trafficking. These micron-sized co-assemblies directly enter the cytosol and then release mRNA, bypassing conventional pathways and thus avoiding the lysosomal degradation. This simple approach involving short oligopeptides and salt addition to create optimal micron-sized co-assembly with mRNA should open new avenues to overcome endosomal barriers for intracellular delivery of nucleic acid therapeutics.
Collapse
Affiliation(s)
- Ruilu Feng
- Chemical and Biological Engineering Department, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
| | - Mehrnoosh Rafiei
- Chemical and Biological Engineering Department, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
| | - Kalindu S Fernando
- Chemical and Biological Engineering Department, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
| | - Ying Chau
- Chemical and Biological Engineering Department, The Hong Kong University of Science and Technology, Hong Kong SAR, China.
| |
Collapse
|