1
|
Wang TY, Krylov SN. Deterministic error propagation in ITC: Revealing multi-fold errors in K d values under standard conditions. Biophys Chem 2025; 323:107455. [PMID: 40349382 DOI: 10.1016/j.bpc.2025.107455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 05/05/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Accurate determination of the equilibrium dissociation constant (Kd) is essential in fields such as drug discovery and molecular diagnostics, where a rigorous understanding of molecular interactions drives critical decisions. Among established techniques, isothermal titration calorimetry (ITC) is highly valued for its ability to directly capture binding thermodynamics without the need for labeling or immobilization. However, while ITC is often praised for its precision, potential inaccuracies due to the systematic errors in experimental variables (analyte concentrations and measured heat) are frequently overlooked. One key reason for this oversight is the lack of a deterministic framework that explicitly demonstrates how ITC-derived Kd values can be affected by these errors. To address this gap, we derived a closed-form mathematical model for error propagation in ITC-based Kd determination, quantifying the impact of inaccuracies in analyte concentrations and measured heat on the resulting Kd. This framework provides a robust foundation for understanding and predicting the influence of these systematic errors on Kd accuracy. Using this solution, we demonstrate that even within the conventionally recommended c-value range of 10-100, expected systematic errors in concentrations and heat can potentially lead to significant multi-fold deviations in Kd. These findings underscore the need for quantitative accuracy assessments in ITC experiments and highlight the importance of developing practical tools to support such evaluations.
Collapse
Affiliation(s)
- Tong Ye Wang
- Department of Chemistry, York University, 4700 Keele St, Toronto, ON M3J 1P3, Canada; Centre for Research on Biomolecular Interactions, York University, 4700 Keele St, Toronto, ON M3J 1P3, Canada
| | - Sergey N Krylov
- Department of Chemistry, York University, 4700 Keele St, Toronto, ON M3J 1P3, Canada; Centre for Research on Biomolecular Interactions, York University, 4700 Keele St, Toronto, ON M3J 1P3, Canada.
| |
Collapse
|
2
|
Khaled G, Benvegnu T, Amin K, Tranchimand S, Chamieh H. Glycosyltransferase enzymatic assays: Overview and comparative analysis. Anal Biochem 2025; 702:115826. [PMID: 40049438 DOI: 10.1016/j.ab.2025.115826] [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: 09/09/2024] [Revised: 03/01/2025] [Accepted: 03/01/2025] [Indexed: 03/10/2025]
Abstract
Glycosyltransferases (GTs) are enzymes that catalyze the transfer of an activated sugar donor to a variety of acceptors including proteins, lipids, carbohydrates, and other small molecules. GTs participate in numerous cellular and physiological processes in both prokaryotic and eukaryotic cells. Those include prokaryotic cell wall biogenesis, eukaryotic post-translational protein modifications, extracellular matrix synthesis, cell signaling, biofilm formation and many others. As such, GTs are exploited as molecular therapeutical targets but also as synthetic tools for the development of polysaccharides and glycoconjugates. In vitro study of GTs activities is now essential to characterize the growing number of predicted GTs, available from sequenced genomes, in order to determine their specificities, their modes of action and their roles in vivo. However, characterization of glycosyltransferases in vitro, both on cellular extracts and on purified enzymes, faces significant challenges. Many methods are currently employed i. e. radiochemical techniques, spectrometric measurements, generally after coupling with∗ other reactions, and even more sophisticated strategies involving product separations by chromatography or/and electrophoresis, followed by detailed structural analysis by NMR or mass spectrometry. Here we overview the common methods deployed for the characterization of GTs. We highlight the challenges arising from these enzymes. The advantages and limitations of each of the presented techniques are also discussed.
Collapse
Affiliation(s)
- Ghazal Khaled
- Univ Rennes, Ecole Nationale Supérieure de Chimie de Rennes, CNRS, ISCR-UMR 6226, F-35000, Rennes, France; Lebanese University, EDST, Azm Center for Research in Biotechnology and Its Applications, LBA3B, Tripoli, Lebanon
| | - Thierry Benvegnu
- Univ Rennes, Ecole Nationale Supérieure de Chimie de Rennes, CNRS, ISCR-UMR 6226, F-35000, Rennes, France
| | - Khadija Amin
- Univ Rennes, Ecole Nationale Supérieure de Chimie de Rennes, CNRS, ISCR-UMR 6226, F-35000, Rennes, France; Lebanese University, EDST, Azm Center for Research in Biotechnology and Its Applications, LBA3B, Tripoli, Lebanon
| | - Sylvain Tranchimand
- Univ Rennes, Ecole Nationale Supérieure de Chimie de Rennes, CNRS, ISCR-UMR 6226, F-35000, Rennes, France.
| | - Hala Chamieh
- Lebanese University, EDST, Azm Center for Research in Biotechnology and Its Applications, LBA3B, Tripoli, Lebanon; Lebanese University, Faculty of Science, Rafic Hariri Campus, Hadat, Lebanon.
| |
Collapse
|
3
|
Vidal FX, Gil J, Gregson M, Zeder-Lutz G, Hideux M, Lemoine J, Krimm I, Wagner R, Dugas V, Demesmay C. Development of ultra-miniaturized weak affinity chromatography coupled to mass spectrometry as a high throughput fragment screening method against wild-type and purified membrane proteins embedded in biomimetic membranes. Anal Chim Acta 2025; 1353:343950. [PMID: 40221197 DOI: 10.1016/j.aca.2025.343950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Membrane proteins, which make up approximately 30 % of the proteome, are important drug targets but present many challenges in drug discovery, including limited production rates, low final yields of pure and functionally folded proteins, and instability in aqueous media. The problems encountered with membrane proteins are even more critical in the Fragment Based Drug Discovery, where the discovery of potential drug candidates is hampered by the limited availability of efficient methods for rapid screening of weak fragment-protein interactions. RESULTS In this work, we propose the coupling of miniaturized weak affinity chromatography with mass spectrometry (nano-WAC-MS) as an innovative strategy for the rapid screening of fragments capable of weak binding to a selected membrane protein. An integral membrane protein (AA2AR) was incorporated into biotinylated nanodiscs, which were subsequently immobilized on a miniaturized monolithic streptavidin column (75 μm i.d., 300 nL volume). The coupling of these miniaturized affinity columns (each consuming less than 1 μg of protein) to mass spectrometry (MRM mode) has been optimized to maximize the low affinity range and increase throughput so that 150 fragments can be injected in a single analysis, with a DMSO content as high as 10 %, with no influence on the affinity. Hits are identified by comparing their retention with that measured on control columns prepared with empty nanodiscs. SIGNIFICANCE The results of this screening are compared with those obtained by NMR and newly identified hits are confirmed by either competition experiments or frontal affinity experiments. We show that this nanodisc-based strategy, which provides a stable and native-like lipid environment for the protein (columns can be used for several days), should also work with other membrane proteins embedded in nanodiscs.
Collapse
Affiliation(s)
- François-Xavier Vidal
- Universite Claude Bernard Lyon1, ISA, UMR5280, CNRS, 5 rue de la Doua, Villeurbanne, 69100, France
| | - Julie Gil
- Universite Claude Bernard Lyon1, ISA, UMR5280, CNRS, 5 rue de la Doua, Villeurbanne, 69100, France
| | - Maud Gregson
- Universite Claude Bernard Lyon1, ISA, UMR5280, CNRS, 5 rue de la Doua, Villeurbanne, 69100, France
| | - Gabrielle Zeder-Lutz
- Plateforme IMPReSs, CNRS UAR3286, Plateforme de chimie biologique intégrative de Strasbourg, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Maria Hideux
- Institut de recherche et développement Servier Paris-Saclay-22, route 128, 91190 Gif sur Yvette, France
| | - Jérôme Lemoine
- Universite Claude Bernard Lyon1, ISA, UMR5280, CNRS, 5 rue de la Doua, Villeurbanne, 69100, France
| | - Isabelle Krimm
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon L, 69373, France
| | - Renaud Wagner
- Plateforme IMPReSs, CNRS UAR3286, Plateforme de chimie biologique intégrative de Strasbourg, Ecole Supérieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Vincent Dugas
- Universite Claude Bernard Lyon1, ISA, UMR5280, CNRS, 5 rue de la Doua, Villeurbanne, 69100, France
| | - Claire Demesmay
- Universite Claude Bernard Lyon1, ISA, UMR5280, CNRS, 5 rue de la Doua, Villeurbanne, 69100, France.
| |
Collapse
|
4
|
Gruca-Stryjak K, Maciak K, Winiewska-Szajewska M, Jurkiewicz A, Gora M, Kacprzak MM, Drgas O, Bialek-Proscinska A, Sobczynska-Tomaszewska A, Pluta KD, Jamsheer A, Markwitz W, Poznanski J, Burzynska B. A novel NEK1 variant disturbs the interaction between the C-terminal fragment of NEK1 and the VDAC1 channel, causing lethal short-rib polydactyly syndrome. Bone 2025; 195:117471. [PMID: 40147672 DOI: 10.1016/j.bone.2025.117471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/07/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
The NIMA-related kinase 1 (NEK1) gene belongs to the Never in Mitosis Gene A (NIMA) kinase family, a group whose members play essential roles in cell cycle regulation, specifically in cell division and ciliogenesis. Mutations in the NEK1 gene have been implicated in several diseases, including short-rib polydactyly syndrome (SRPS). SRPS is a bone growth disorder characterized by severe congenital anomalies. Here, we describe a family with a lethal form of SRPS due to a novel intronic variant in the NEK1 gene. Basing on whole-exome sequencing of fetuses with SRPS we identified a homozygous variant of the NEK1 gene at position c.3584-10T>A as the causative mutation. Bioinformatic methods and minigene splicing assays were then used to assess the harmfulness and functional impact of the variant. We found that the identified mutation leads to the synthesis of the NEK1 protein lacking 90C-terminal residues following the last coiled-coil region. Additional experiments were performed to identify proteins that interact with the C-terminal fragment of NEK1 absent in the mutated protein. We suggest that the interaction between the C-terminal fragment of NEK1 and the VDAC1 channel is essential for the VDAC1 phosphorylation, the absence of which is likely to affect ciliogenesis.
Collapse
Affiliation(s)
- Karolina Gruca-Stryjak
- Department of Perinatology, Poznan University of Medical Sciences, Poznan, Poland; Centers of Medical Genetics GENESIS, Poznan, Poland
| | - Karolina Maciak
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | - Aneta Jurkiewicz
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Monika Gora
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | | | | | | | - Krzysztof D Pluta
- Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Aleksander Jamsheer
- Department of Medical Genetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Wieslaw Markwitz
- Department of Perinatology, Poznan University of Medical Sciences, Poznan, Poland
| | - Jaroslaw Poznanski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Beata Burzynska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
5
|
Wu X, Gong X, Xie T. Mechanisms of aureobasidin A inhibition and drug resistance in a fungal IPC synthase complex. Nat Commun 2025; 16:5010. [PMID: 40442105 PMCID: PMC12122681 DOI: 10.1038/s41467-025-60423-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 05/22/2025] [Indexed: 06/02/2025] Open
Abstract
The enzyme inositol phosphorylceramide (IPC) synthase is essential for survival and virulence in fungi, while absent in mammals, thus representing a potential target for antifungal treatments. Aureobasidin A (AbA), a natural cyclic peptide, displays antifungal activity and inhibits IPC synthase, but the precise molecular mechanism remains unclear. Here, we present the cryo-EM structure of the Saccharomyces cerevisiae IPC synthase, composed of catalytic subunit Aur1 and regulatory subunit Kei1, in its AbA-bound state. The complex is resolved as a dimer of Aur1-Kei1 heterodimers, with Aur1 mediating homodimerization. AbA occupies a predominantly hydrophobic pocket in the catalytic core domain of each Aur1 subunit, blocking the entry of both substrates. Mutations conferring AbA resistance cluster near the AbA-binding site, thus interfering with AbA binding. Our study lays a foundation for the development of therapeutic drugs targeting fungal IPC synthase.
Collapse
Affiliation(s)
- Xinyue Wu
- Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Xin Gong
- Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
| | - Tian Xie
- Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
6
|
Sołtys K, Skowronek K, Bystranowska D, Wycisk K, Ożyhar A. The intrinsically disordered AB region: a key modulator of the molecular properties of human RXRγ. Cell Commun Signal 2025; 23:243. [PMID: 40420113 PMCID: PMC12105186 DOI: 10.1186/s12964-025-02247-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 05/12/2025] [Indexed: 05/28/2025] Open
Abstract
The human retinoid X receptor γ (hRXRγ) is one of three characterized RXR subtypes, transcription factors belonging to the nuclear receptor superfamily. All RXR subtypes share nearly identical structural elements, including a conserved DNA-binding domain, a D region, a ligand-binding domain, and an F region. However, each subtype possesses a unique N-terminal AB region, which modulates the transcriptional activation of target genes in a cell- and promoter-dependent manner through its ligand-independent activation function involved in protein-protein interactions. Despite the functional significance of the AB region, its structural contributions, particularly in the context of the full-length receptor, remain largely unexplored. Here, we uncover the role of the AB region of hRXRγ in modulating the molecular properties of the receptor. A comparative analysis of the full-length receptor (hRXRγ) and a deletion mutant lacking the AB region (ΔABhRXRγ) highlights the critical role of the intrinsically disordered AB region in modulating the structural and functional properties of hRXRγ, including its ability to oligomerize, its overall stability, and conformation heterogeneity. The AB region does not act as an independent unit but amalgamates with the rest of the receptor, which fine-tunes the structural variability of hRXRγ, making it responsive to environmental conditions. These findings highlight the AB region as a critical determinant of hRXRγ's structural features and, potentially, its transcriptional potential.
Collapse
Affiliation(s)
- Katarzyna Sołtys
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, Wrocław, 50-370, Poland.
| | - Krzysztof Skowronek
- International Institute of Molecular and Cell Biology, 4 Ks. Trojdena Street, Warsaw, 02-109, Poland
| | - Dominika Bystranowska
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, Wrocław, 50-370, Poland
| | - Krzysztof Wycisk
- International Institute of Molecular and Cell Biology, 4 Ks. Trojdena Street, Warsaw, 02-109, Poland
| | - Andrzej Ożyhar
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wrocław University of Science and Technology, Wybrzeże Wyspiańskiego 27, Wrocław, 50-370, Poland
| |
Collapse
|
7
|
Qin L, Hu C, Zhao Q, Wang Y, Fan D, Lin A, Xiang L, Chen Y, Shao J. Unraveling the role of Ctla-4 in intestinal immune homeostasis through a novel Zebrafish model of inflammatory bowel disease. eLife 2025; 13:RP101932. [PMID: 40392591 PMCID: PMC12092003 DOI: 10.7554/elife.101932] [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] [Indexed: 05/22/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic and relapsing immune-mediated disorder characterized by intestinal inflammation and epithelial injury. The underlying causes of IBD are not fully understood, but genetic factors have been implicated in genome-wide association studies, including CTLA-4, an essential negative regulator of T cell activation. However, establishing a direct link between CTLA-4 and IBD has been challenging due to the early lethality of CTLA-4 knockout mice. In this study, we identified zebrafish Ctla-4 homolog and investigated its role in maintaining intestinal immune homeostasis by generating a Ctla-4-deficient (ctla-4-/-) zebrafish line. These mutant zebrafish exhibited reduced weight, along with impaired epithelial barrier integrity and lymphocytic infiltration in their intestines. Transcriptomics analysis revealed upregulation of inflammation-related genes, disturbing immune system homeostasis. Moreover, single-cell RNA-sequencing analysis indicated increased Th2 cells and interleukin 13 expression, along with decreased innate lymphoid cells and upregulated proinflammatory cytokines. Additionally, Ctla-4-deficient zebrafish exhibited reduced diversity and an altered composition of the intestinal microbiota. All these phenotypes closely resemble those found in mammalian IBD. Lastly, supplementation with Ctla-4-Ig successfully alleviated intestinal inflammation in these mutants. Altogether, our findings demonstrate the pivotal role of Ctla-4 in maintaining intestinal homeostasis. Additionally, they offer substantial evidence linking CTLA-4 to IBD and establish a novel zebrafish model for investigating both the pathogenesis and potential treatments.
Collapse
Affiliation(s)
- Lulu Qin
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Chongbin Hu
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Qiong Zhao
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Yong Wang
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Dongdong Fan
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Aifu Lin
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Lixin Xiang
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
| | - Ye Chen
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
- Department of Genetic and Metabolic Disease, the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child HealthHangzhouChina
| | - Jianzhong Shao
- College of Life Sciences, Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Zhejiang UniversityHangzhouChina
- Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and TechnologyQingdaoChina
| |
Collapse
|
8
|
Yang Z, Wei H, Gan Y, Liu H, Cao Y, An H, Que X, Gao Y, Zhu L, Tan S, Liu X, Sun L. Structural insights into auxin influx mediated by the Arabidopsis AUX1. Cell 2025:S0092-8674(25)00463-5. [PMID: 40378849 DOI: 10.1016/j.cell.2025.04.028] [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: 09/27/2024] [Revised: 02/28/2025] [Accepted: 04/21/2025] [Indexed: 05/19/2025]
Abstract
Auxin is crucial in orchestrating diverse aspects of plant growth and development and modulating responses to environmental signals. The asymmetric spatiotemporal distribution of auxin generates local gradient patterns, which are regulated by both cellular auxin influx and efflux. The AUXIN1/LIKE-AUX1 (AUX1/LAX) family transporters have been identified as major auxin influx carriers. Here, we characterize the auxin uptake mediated by AUX1 from Arabidopsis thaliana. Using cryoelectron microscopy (cryo-EM), we determine its structure in three states: the auxin-unbound, the auxin-bound, and the competitive inhibitor, 3-chloro-4-hydroxyphenylacetic acid (CHPAA)-bound state. All structures adopt an inward-facing conformation. In the auxin-bound structure, indole-3-acetic acid (IAA) is coordinated to AUX1 primarily through hydrogen bonds with its carboxyl group. The functional roles of key residues in IAA binding are validated by in vitro and in planta analyses. CHPAA binds to the same site as IAA. These findings advance our understanding of auxin transport in plants.
Collapse
Affiliation(s)
- Zhisen Yang
- Department of Neurology of The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Hong Wei
- Department of Neurology of The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Yulin Gan
- Department of Neurology of The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Huihui Liu
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Yang Cao
- Department of Neurology of The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Huihui An
- Department of Neurology of The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Xiuzheng Que
- Department of Neurology of The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Yongxiang Gao
- Department of Neurology of The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Lizhe Zhu
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Shutang Tan
- Department of Neurology of The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230027, China.
| | - Xin Liu
- Department of Neurology of The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230027, China.
| | - Linfeng Sun
- Department of Neurology of The First Affiliated Hospital of USTC, MOE Key Laboratory for Membraneless Organelles and Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China; Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei 230027, China.
| |
Collapse
|
9
|
Kim K, Chung KY. Protocol for measuring β-arrestin1-phosphorylated peptide interaction and analyzing conformational dynamics of β-arrestin1. STAR Protoc 2025; 6:103823. [PMID: 40347475 DOI: 10.1016/j.xpro.2025.103823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/20/2025] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
Arrestins associate with phosphorylated G protein-coupled receptors and undergo conformational changes. Here, we present a protocol for measuring β-arrestin1-phosphorylated peptide interaction and analyzing conformational dynamics of β-arrestin1. We describe steps for constructing expression plasmids, expressing and purifying β-arrestin1, and performing hydrogen/deuterium exchange mass spectrometry analysis of V2Rpp-β-arrestin1 complex. We then detail procedures for measuring binding affinity using microscale thermophoresis. This protocol has potential application in analyzing conformational dynamics and measuring binding affinity of arrestins. For complete details on the use and execution of this protocol, please refer to Kim et al.1.
Collapse
Affiliation(s)
- Kiae Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ka Young Chung
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
10
|
Lei Q, Yang H, Wu SX, Xu L, Wei D, Wang JJ, Jiang HB. BdorOBP32 Perceiving β-Caryophyllene: A Molecular Target for Female Attractant Development in Bactrocera dorsalis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:11209-11217. [PMID: 40272313 DOI: 10.1021/acs.jafc.5c00167] [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: 04/25/2025]
Abstract
Bactrocera dorsalis is a destructive agricultural pest that attacks over 600 plant species. β-Caryophyllene is considered a potential compound for developing novel female attractants due to its attraction to B. dorsalis females. However, the unknown perception mechanism of β-caryophyllene has been the bottleneck of this process. Odorant binding proteins (OBPs) function to bind odorants and transport them to olfactory receptors. Here, behavioral assays revealed that β-caryophyllene strongly attracted mated, instead of virgin females. RT-qPCR confirmed BdorOBP32 up-regulation out of five OBPs in mated females compared to virgin females. Microscale thermophoresis (MST) results showed BdorOBP32 bind β-caryophyllene with relatively high affinity. Subsequently, CRISPR/Cas9 knockout of BdorOBP32 reduced electroantennograms responses and behavioral preferences to β-caryophyllene in mutants. Moreover, molecular docking and behavioral analysis identified a novel female attractant (α-angelica lactone) targeting BdorOBP32. These findings highlight BdorOBP32 plays critical roles in β-caryophyllene perception and offer new insights for developing novel olfactory behavior modulators.
Collapse
Affiliation(s)
- Quan Lei
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing 400715, China
| | - Hui Yang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing 400715, China
| | - Shuang-Xiong Wu
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing 400715, China
| | - Li Xu
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing 400715, China
| | - Dong Wei
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing 400715, China
| | - Jin-Jun Wang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing 400715, China
| | - Hong-Bo Jiang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing 400716, China
- Key Laboratory of Agricultural Biosafety and Green Production of Upper Yangtze River (Ministry of Education), Southwest University, Chongqing 400715, China
| |
Collapse
|
11
|
Siddiq A A, Dileep SA, Sj AR, Singam SSR, Martin A. Saffron and its active constituents ameliorate hypercholesterolemia by inhibiting PCSK9 and modulating Sortilin, LDLR, and SREBP-2 signaling in high fat diet induced hypercholesterolemic C57BL/6 mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119697. [PMID: 40157403 DOI: 10.1016/j.jep.2025.119697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 03/17/2025] [Accepted: 03/23/2025] [Indexed: 04/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Saffron (Crocus sativus L.) has long been used in Ayurveda, Iranian, and Chinese traditional medicine as a natural remedy for hypercholesterolemia, obesity, and liver disorders though its therapeutic mechanism remains unclear. AIM OF THE STUDY This study explores the mechanism by which saffron extract (SE), crocin (CN), and crocetin (CR) mitigate high fat diet (HFD) induced hypercholesterolemia and hepatic inflammation in C57BL/6 mice, focusing on their inhibition of proprotein convertase subtilisin/kexin type 9 (PCSK9). MATERIALS AND METHODS C57BL/6 mice (N = 10/group) were fed either a, normal diet, HFD, or HFD supplemented with SE, CN, CR, or atorvastatin for 12 weeks. Plasma lipids and inflammatory markers were measured. Histopathological changes were assessed via H&E and Sudan black staining. Gene expression was analyzed using qRT-PCR, and ligand-protein interactions were studied using molecular docking, simulation, and thermophoresis. RESULTS HFD-fed mice exhibited dyslipidemia, liver damage, and inflammation, which SE, CN, and CR significantly improved. Treatments reduced cholesterol, triglycerides, and reactive oxygen species, reversed fatty liver degeneration, and downregulated PCSK9 and sortilin expression while upregulating LDLR. They suppressed transcription factors SREBP-1C and SREBP-2 and reduced inflammatory markers, including TNF-α, while increasing IL-10 expression. CR reduced plasma PCSK9 secretion by 39.9 % (p < 0.05). Docking and simulation studies confirmed the strong binding potential of CR and CN to PCSK9. CONCLUSION Saffron and its active components (CN and CR) are novel natural PCSK9 inhibitors that effectively ameliorate hypercholesterolemia by modulating sortilin, LDLR and SREBP-2 pathway, potentially opening the way for developing new therapeutic approaches for managing cholesterol related disorders.
Collapse
Affiliation(s)
- Aisha Siddiq A
- Department of Biochemistry, CSIR - Central Food Technological Research Institute, Mysuru, 570 020, Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201 002, India.
| | - Shaik Abdul Dileep
- Food Safety and Analytical Quality Control Laboratory, CSIR - Central Food Technological Research Institute, Mysuru, 570 020, Karnataka, India.
| | - Aditya Rao Sj
- Plant Cell Biotechnology Department, CSIR - Central Food Technological Research Institute, Mysuru, 570 020, Karnataka, India.
| | - Siva Sankara Reddy Singam
- Food Safety and Analytical Quality Control Laboratory, CSIR - Central Food Technological Research Institute, Mysuru, 570 020, Karnataka, India.
| | - Asha Martin
- Department of Biochemistry, CSIR - Central Food Technological Research Institute, Mysuru, 570 020, Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201 002, India.
| |
Collapse
|
12
|
Dettmer SJ, Williams HD, Napier R, Beames JM, Walker-Griffiths S, Craggs TD, Hannon MJ. Supramolecular Recognition of a DNA Four-Way Junction by an M 2L 4 Metallo-Cage, Inspired by a Simulation-Guided Design Approach. Angew Chem Int Ed Engl 2025:e202504866. [PMID: 40243103 DOI: 10.1002/anie.202504866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 04/18/2025]
Abstract
DNA four-way junctions (4WJs) play an important biological role in DNA repair and recombination, and viral regulation, and are attractive therapeutic targets. Compounds that recognise the junction structure are rare; in this work, we describe cationic metallo-supramolecular M2L4 cages as a new type of 4WJ binder with nanomolar affinities. A combination of molecular dynamics (MD) simulations and biophysical experiments show that the size and shape of a compound comprising square planar Pd or Pt and anthracene-based ligands is an excellent fit for the 4WJ cavity. Whilst the cage is also capable of binding to three-way junctions (3WJs) and Y-fork structures, we show that the 4WJ is the preferred DNA target, and that duplex B-DNA is not a competitor. Among 3WJs, T-shape bulged 3WJs are bound more preferably than perfect Y-shaped 3WJs. Whilst previous work studying M2L4 metallo-supramolecular cages has focused on binding inside their structures, this work exploits the external aromatic surfaces of the supramolecule, creating a supramolecular guest that ideally matches the DNA host cavity. This approach allows available structures to be identified as potential junction binders and then screened for their fit to a nucleic acid junction target using simulations. This has potential to significantly accelerate discovery.
Collapse
Affiliation(s)
- Samuel J Dettmer
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Hugo D Williams
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Richard Napier
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Joseph M Beames
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | | | - Timothy D Craggs
- School of Mathematical and Physical Sciences, Centre for Single-Molecule Biology, University of Sheffield, Sheffield, S3 7HF, UK
| | - Michael J Hannon
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| |
Collapse
|
13
|
Zhou J, Ridderbeek K, Zou P, Naden AB, Gaussmann S, Song F, Falter-Braun P, Kay ER, Sattler M, Cui J. Modular Nanoparticle Platform for Solution-Phase Optical Sensing of Protein-Protein Interactions. ACS APPLIED OPTICAL MATERIALS 2025; 3:676-688. [PMID: 40176919 PMCID: PMC11959585 DOI: 10.1021/acsaom.4c00486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 04/05/2025]
Abstract
Protein-protein interactions regulate essentially all cellular processes. Understanding these interactions, including the quantification of binding parameters, is crucial for unraveling the molecular mechanisms underlying cellular pathways and, ultimately, their roles in cellular physiology and pathology. Current methods for measuring protein-protein interactions in vitro generally require amino acid conjugation of fluorescent tags, complex instrumentation, large amounts of purified protein, or measurement at extended surfaces. Here, we present an elegant nanoparticle-based platform for the optical detection of protein-protein interactions in the solution phase. We synthesized gold-coated silver decahedral nanoparticles possessing high chemical stability and exceptional optical sensing properties. The nanoparticle surface is then tailored for specific binding to commonly used polyhistidine tags of recombinant proteins. Sequential addition of proteins to the nanoparticle suspension results in spectral shifts of the localized surface plasmon resonance that can be monitored by conventional UV-vis spectrophotometry. With this approach, we demonstrate both the qualitative detection of specific protein-protein interactions and the quantification of equilibrium and kinetic binding parameters between small globular proteins. Requiring minimal protein quantities and basic laboratory equipment, this technique offers a simple, economical, and modular approach to characterizing protein-protein interactions, holds promise for broad use in future studies, and may serve as a template for future biosensing technologies.
Collapse
Affiliation(s)
- Jieying Zhou
- Helmholtz
Pioneer Campus, Helmholtz Munich, Neuherberg 85764, Germany
| | | | - Peijian Zou
- Institute
of Structural Biology, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg 85764, Germany
- Bavarian
NMR Center, Department of Bioscience, School of Natural Sciences, Technical University of Munich, Garching 85748, Germany
| | - Aaron B. Naden
- EaStCHEM
School of Chemistry, University of St. Andrews, St. Andrews KY16 9ST, U.K.
| | - Stefan Gaussmann
- Institute
of Structural Biology, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg 85764, Germany
- Bavarian
NMR Center, Department of Bioscience, School of Natural Sciences, Technical University of Munich, Garching 85748, Germany
| | - Fangyuan Song
- Helmholtz
Pioneer Campus, Helmholtz Munich, Neuherberg 85764, Germany
| | - Pascal Falter-Braun
- Institute
of Network Biology (INET), Molecular Targets and Therapeutics Center
(MTTC), Helmholtz Munich, Neuherberg 85764 Germany
- Microbe-Host
Interactions, Faculty of Biology, Ludwig-Maximilians-Universität
(LMU) München, Planegg-Martinsried 82152, Germany
| | - Euan R. Kay
- EaStCHEM
School of Chemistry, University of St. Andrews, St. Andrews KY16 9ST, U.K.
| | - Michael Sattler
- Institute
of Structural Biology, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg 85764, Germany
- Bavarian
NMR Center, Department of Bioscience, School of Natural Sciences, Technical University of Munich, Garching 85748, Germany
| | - Jian Cui
- Helmholtz
Pioneer Campus, Helmholtz Munich, Neuherberg 85764, Germany
- Department
of Bioscience, School of Natural Sciences, Technical University of Munich, Garching 85748, Germany
| |
Collapse
|
14
|
Nguyen E, Agbavor C, Steenhaut A, Pratyush MR, Hiller NL, Cahoon LA, Mikheyeva IV, Ng WL, Bridges AA. A small periplasmic protein governs broad physiological adaptations in Vibrio cholerae via regulation of the DbfRS two-component system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.645060. [PMID: 40196685 PMCID: PMC11974885 DOI: 10.1101/2025.03.24.645060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Two-component signaling pathways allow bacteria to sense and respond to environmental changes, yet the sensory mechanisms of many remain poorly understood. In the pathogen Vibrio cholerae, the DbfRS two-component system controls the biofilm lifecycle, a critical process for environmental persistence and host colonization. Here, we identified DbfQ, a small periplasmic protein encoded adjacent to dbfRS, as a direct modulator of pathway activity. DbfQ directly binds the sensory domain of the histidine kinase DbfS, shifting it toward phosphatase activity and promoting biofilm dispersal. In contrast, outer membrane perturbations, caused by mutations in lipopolysaccharide biosynthesis genes or membrane-damaging antimicrobials, activate phosphorylation of the response regulator DbfR. Transcriptomic analyses reveal that DbfR phosphorylation leads to broad transcriptional changes spanning genes involved in biofilm formation, central metabolism, peptidoglycan synthesis, and cellular stress responses. Constitutive DbfR phosphorylation imposes severe fitness costs in an infection model, highlighting this pathway as a potential target for anti-infective therapeutics. We find that dbfQRS-like genetic modules are widely present across bacterial phyla, underscoring their broad relevance in bacterial physiology. Collectively, these findings establish DbfQ as a new class of periplasmic regulator that influences two-component signaling and bacterial adaptation.
Collapse
Affiliation(s)
- Emmy Nguyen
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Charles Agbavor
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Anjali Steenhaut
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - M R Pratyush
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - N. Luisa Hiller
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Laty A. Cahoon
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Irina V. Mikheyeva
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Wai-Leung Ng
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Andrew A. Bridges
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
15
|
Mastalipour M, Gering I, Coronado MA, González JEH, Willbold D, Eberle RJ. Novel peptide inhibitor for the Chikungunya virus nsP2 protease: Identification and characterization. CURRENT RESEARCH IN MICROBIAL SCIENCES 2025; 8:100376. [PMID: 40165933 PMCID: PMC11957602 DOI: 10.1016/j.crmicr.2025.100376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Chikungunya virus (CHIKV) is an emerging pathogen affecting populations worldwide, with rapidly increasing infection rates. CHIKV, an arbovirus of the alphavirus genus, is predominantly found in tropical regions and transmitted by Aedes mosquitoes. Climate change has accelerated the global spread of these vectors, leading to outbreaks in non-tropical regions, including parts of Europe. The absence of antiviral therapies and the potential for co-infections with other viruses make CHIKV a significant public health concern. CHIKV replication relies on nsP2 cysteine protease activity to cleave its viral polyprotein into functional nonstructural and structural proteins. Targeting the nsP2 protease represents a promising strategy for antiviral therapy development. In this study, phage display was used to screen a library of peptides for potential binders of the target protease. Biophysical and biochemical analyses of the identified peptides assessed their inhibitory potential. Among the six identified peptides (named as P1-P6), four demonstrated inhibitory effects on the nsP2 protease (nsP2pro). Peptide P1 exhibited the strongest inhibitory effect, with a half-maximal inhibitory concentration (IC50) of 4.6 ± 1.9 µM, and a low cytotoxicity. The secondary structure analysis through CD spectroscopy and homology modelling revealed that P1 adopts an alpha-helical conformation. Finally, molecular dynamics simulations enabled us to investigate the dynamics of the nsP2pro active site and molecular docking was employed to predict the orthosteric binding mode of P1, providing insights into protein-peptide interaction. These findings underscore the potential of peptide P1 as a lead compound for further investigation in the context of CHIKV research.
Collapse
Affiliation(s)
- Mohammadamin Mastalipour
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Ian Gering
- Institut für Biologische Informationsprozesse, Strukturbiochemie (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Mônika Aparecida Coronado
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
- Institut für Biologische Informationsprozesse, Strukturbiochemie (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany
| | | | - Dieter Willbold
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
- Institut für Biologische Informationsprozesse, Strukturbiochemie (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Raphael Josef Eberle
- Institut für Biochemische Pflanzenphysiologie, Heinrich-Heine-Universität Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| |
Collapse
|
16
|
Teichmann T, Pflüger-Müller B, Giménez VMM, Sailer F, Dirks H, Zehr S, Warwick T, Brettner F, Munoz-Tello P, Zimmer A, Tegeder I, Thomas D, Gurke R, Günther S, Heering J, Proschak E, Geisslinger G, Bibli IS, zu Heringdorf DM, Manucha W, Windbergs M, Knapp S, Weigert A, Leisegang MS, Kojetin D, Brandes RP. The endocannabinoid anandamide mediates anti-inflammatory effects through activation of NR4A nuclear receptors. Br J Pharmacol 2025; 182:1164-1182. [PMID: 39563075 PMCID: PMC11790392 DOI: 10.1111/bph.17366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND AND PURPOSE Endocannabinoids are lipid mediators, which elicit complex biological effects that extend beyond the central nervous system. Tissue concentrations of endocannabinoids increase in atherosclerosis, and for the endocannabinoid N-arachidonoyl-ethanolamine (anandamide, AEA), this has been linked to an anti-inflammatory function. In this study, we set out to determine the anti-inflammatory mechanism of action of AEA, specifically focusing on vascular smooth muscle cells. EXPERIMENTAL APPROACH RNA-sequencing, RT-qPCR, LC-MS/MS, NanoBit, ChIP, microscale thermophoresis, NMR structural footprinting, Gal4 reporter gene assays and loss of function approaches in cell and ex vivo organ culture were used. KEY RESULTS AEA pretreatment attenuated the cytokine-mediated induction of inflammatory gene expression such as CCL2. This effect was also observed in preparations obtained from cannabinoid receptor knockout mice and after pertussis toxin treatment. The anti-inflammatory effect of AEA required preincubation, suggesting an effect through gene induction. AEA increased the expression of the nuclear receptors NR4A1 and NR4A2. Knockdown and knockout of these receptors blocked the AEA-mediated anti-inflammatory effect in cell culture and aortic organ culture, respectively. Conversely, NR4A agonists (CsnB, C-DIM12) attenuated inflammatory gene expression. AEA binds to NR4A, and mutations in NR4A attenuated this effect. The interaction of AEA with NR4A caused recruitment of the nuclear corepressor NCoR1 to the CCL2 promoter, resulting in gene suppression. CONCLUSION AND IMPLICATIONS By binding to NR4A, AEA elicits an anti-inflammatory response in vascular smooth muscle cells. NR4A-binding by AEA analogues may represent novel anti-inflammatory agents.
Collapse
MESH Headings
- Endocannabinoids/pharmacology
- Animals
- Arachidonic Acids/pharmacology
- Anti-Inflammatory Agents/pharmacology
- Polyunsaturated Alkamides/pharmacology
- Mice
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 1/agonists
- Mice, Knockout
- Humans
- Mice, Inbred C57BL
- Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 2/genetics
- Male
- Cells, Cultured
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
Collapse
Affiliation(s)
- Tom Teichmann
- Goethe University Frankfurt, Institute for Cardiovascular Physiology, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Beatrice Pflüger-Müller
- Goethe University Frankfurt, Institute for Cardiovascular Physiology, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Virna Margarita Martín Giménez
- Universidad Católica de Cuyo, Instituto de Investigaciones en Ciencias Químicas, Facultad de Ciencias Químicas y Tecnológicas, San Juan, Argentina
| | - Fiona Sailer
- Goethe University Frankfurt, Institute of Biochemistry I, Faculty of Medicine, Frankfurt, Germany
| | - Henrik Dirks
- Goethe University Frankfurt, Institute for Cardiovascular Physiology, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Simonida Zehr
- Goethe University Frankfurt, Institute for Cardiovascular Physiology, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Timothy Warwick
- Goethe University Frankfurt, Institute for Cardiovascular Physiology, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Felix Brettner
- Goethe University Frankfurt, Institute of Pharmaceutical Technology, Frankfurt am Main, Germany
| | - Paola Munoz-Tello
- Vanderbilt University, Department of Biochemistry, Nashville, Tennessee, United States
| | - Andreas Zimmer
- University of Bonn, Institute of Molecular Psychiatry, Medical Faculty, Bonn, Germany
| | - Irmgard Tegeder
- Goethe-University Frankfurt, Institute of Clinical Pharmacology, Medical Faculty, Frankfurt, Germany
| | - Dominique Thomas
- Goethe-University Frankfurt, Institute of Clinical Pharmacology, Medical Faculty, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt, Germany
| | - Robert Gurke
- Goethe-University Frankfurt, Institute of Clinical Pharmacology, Medical Faculty, Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt, Germany
| | - Stefan Günther
- Max-Plank-Institute for Heart- and Lung research (MPI-HLR), Bad Nauheim, Germany
| | - Jan Heering
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt, Germany
| | - Ewgenij Proschak
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMD, Frankfurt, Germany
- Goethe University Frankfurt, Institute of Pharmaceutical Chemistry, Frankfurt, Germany
| | - Gerd Geisslinger
- Goethe-University Frankfurt, Institute of Clinical Pharmacology, Medical Faculty, Frankfurt, Germany
| | - Iris-S. Bibli
- German Centre of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
- Goethe University Frankfurt, Institute for Vascular Signaling, Centre for Molecular Medicine, Frankfurt am Main, Germany
| | - Dagmar Meyer zu Heringdorf
- Goethe University Frankfurt, Institute of General Pharmacology and Toxicology, University Hospital Frankfurt, Frankfurt, Germany
| | - Walter Manucha
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Mendoza 5500, Argentina
- Universidad Nacional de Cuyo, Departamento de Patología, Área de Farmacología, Facultad de Ciencias Médicas, Mendoza 5500, Argentina
| | - Maike Windbergs
- Goethe University Frankfurt, Institute of Pharmaceutical Technology, Frankfurt am Main, Germany
| | - Stefan Knapp
- Goethe University Frankfurt, Institute of Pharmaceutical Chemistry, Frankfurt, Germany
- Goethe University Frankfurt, Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Frankfurt am Main, Germany
| | - Andreas Weigert
- Goethe University Frankfurt, Institute of Biochemistry I, Faculty of Medicine, Frankfurt, Germany
| | - Matthias S. Leisegang
- Goethe University Frankfurt, Institute for Cardiovascular Physiology, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| | - Douglas Kojetin
- Vanderbilt University, Department of Biochemistry, Nashville, Tennessee, United States
- Vanderbilt University, Center for Structural Biology and Institute for Chemical Biology, Nashville, Tennessee, United States
| | - Ralf P. Brandes
- Goethe University Frankfurt, Institute for Cardiovascular Physiology, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt, Germany
| |
Collapse
|
17
|
Ng MY, Wang H, Zhang H, Prucker I, Perera L, Goncharova E, Wamiru A, Jessen HJ, Stanley RE, Shears SB, Luo J, O'Keefe BR, Wilson BAP. Biochemical and biophysical characterization of inositol-tetrakisphosphate 1-kinase inhibitors. J Biol Chem 2025; 301:108274. [PMID: 39922495 PMCID: PMC11927698 DOI: 10.1016/j.jbc.2025.108274] [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: 10/14/2024] [Revised: 01/23/2025] [Accepted: 01/25/2025] [Indexed: 02/10/2025] Open
Abstract
Inositol phosphates (IPs) and inositol pyrophosphate play critical roles in many biological processes such as signaling molecules in pathways responsible for cellular functions involved in growth and maintenance. The biosynthesis of IPs is carried out by a family of inositol phosphate kinases. In mammals, Inositol tetrakisphosphate kinase-1 (ITPK1) phosphorylates inositol-1,3,4-trisphosphate (Ins(1,3,4)P3) and inositol-3,4,5,6-tetrakisphosphate (IP4), generating inositol-1,3,4,5,6-pentakisphosphate (IP5), which can be further phosphorylated to become inositol hexakisphosphate (IP6). ITPK1 also possesses phosphatase activity that can convert IP5 back to IP4; therefore, ITPK1 may serve as a regulatory step in IP6 production. IP6 utilization has been implicated in processes fundamental to cellular sustainability that are severely perturbed in many disease states including RNA editing, DNA repair, chromatin structure organization, and ubiquitin ligation. Therefore, ITPK1, with no known inhibitors in the literature, is a potential molecular target for modulating important processes in several human diseases. By independently coupling ITPK1 phosphatase and kinase activities to luciferase activity, we have developed and used biochemical high-throughput assays to discover eight ITPK1 inhibitors. Further analysis revealed that three of these leads inhibit ITPK1 in an ATP-competitive manner, with low micromolar to nanomolar affinities. We further demonstrate that the most potent ITPK1 inhibitor can regulate cellular ITPK1 activity. We determined the crystal structure of ITPK1 in complex with this inhibitor at a resolution of 2.25 Å. This work provides insight into the design of potential next-generation inhibitors.
Collapse
Affiliation(s)
- Martin Y Ng
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
| | - Huanchen Wang
- Molecular and Cellular Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, USA
| | - Haibo Zhang
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Isabel Prucker
- Institute of Organic Chemistry, University of Freiburg, Freiburg, Germany
| | - Lalith Perera
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, USA
| | - Ekaterina Goncharova
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA; Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Antony Wamiru
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
| | - Henning J Jessen
- Institute of Organic Chemistry, University of Freiburg, Freiburg, Germany; Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Robin E Stanley
- Molecular and Cellular Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, USA
| | - Stephen B Shears
- Molecular and Cellular Biology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina, USA
| | - Ji Luo
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA.
| | - Barry R O'Keefe
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA; Natural Products Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, Maryland, USA.
| | - Brice A P Wilson
- Molecular Targets Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA.
| |
Collapse
|
18
|
Karlowicz A, Dubiel A, Wyszkowska M, Hossain K, Czub J, Szymanski M. Mitochondrial exonuclease EXOG supports DNA integrity by the removal of single-stranded DNA flaps. Nucleic Acids Res 2025; 53:gkaf099. [PMID: 40042814 PMCID: PMC11880807 DOI: 10.1093/nar/gkaf099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 01/27/2025] [Accepted: 02/04/2025] [Indexed: 03/09/2025] Open
Abstract
Single-stranded DNA (ssDNA) is an important intermediate generated during various cellular DNA transactions, primarily during long-patch base excision repair. When displaced by DNA polymerase during strand displacement DNA synthesis, ssDNA forms 5' overhangs (flaps) that are either cleaved by DNA nucleases or protected from degradation upon binding of single-stranded DNA-binding proteins (SSB). Several nucleases are involved in the removal of ssDNA flaps in human mitochondria, namely the endonucleases FEN1 and DNA2, as well as the exonuclease MGME1. In this study, we show that another mitochondrial nuclease, EXOG, cleaves DNA flaps in both free and SSB-protected forms. We established that the presence of the Wing domain in EXOG structure provides additional binding site for ssDNA and 5' flaps irrespective of monovalent salt concentration. Importantly, DNA flap cleavage by EXOG is compatible with the activity of other mitochondrial enzymes involved in DNA replication/repair, e.g. mtSSB, Pol γ, and Lig III, as we were able to reconstitute a multistep reaction of DNA synthesis, flap removal, and nick ligation. Our findings highlight the versatile role of EXOG in maintaining mitochondrial DNA integrity, expanding its DNA processing repertoire to include ssDNA flap removal.
Collapse
Affiliation(s)
- Anna Karlowicz
- Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland
| | - Andrzej B Dubiel
- Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland
| | - Marta Wyszkowska
- Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland
| | - Kazi Amirul Hossain
- Department of Physical Chemistry, Gdansk University of Technology, ul. Narutowicza 11/12, 80-233 Gdansk, Poland
| | - Jacek Czub
- Department of Physical Chemistry, Gdansk University of Technology, ul. Narutowicza 11/12, 80-233 Gdansk, Poland
| | - Michal R Szymanski
- Structural Biology Laboratory, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, University of Gdansk, Abrahama 58, 80-307 Gdansk, Poland
| |
Collapse
|
19
|
Koczurowska A, Carrillo DR, Alai MG, Zakłos-Szyda M, Bujacz G, Pietrzyk-Brzezinska AJ. Structural and biophysical characterization of the cytoplasmic domains of HprS kinase and its interactions with the cognate regulator HprR. Arch Biochem Biophys 2025; 764:110269. [PMID: 39681306 DOI: 10.1016/j.abb.2024.110269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/22/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
The HprSR constitutes the bacterial two-component regulatory system engaged by Escherichia coli to reduce the damaging effects of reactive chlorine and oxygen species present in its cytosol. Hypochlorous acid (HOCl) has been shown to be the molecule capable of activating of the HprSR system. HOCl is produced upon pathogen invasion by phagocytic cells of the human innate immune system, particularly neutrophils, to take advantage of its powerful antimicrobial attributes. Therefore, comprehensive studies concerning bacterial sensing and regulatory HprSR system are indispensable in understanding and effectively eliminating pathogens. Here we present the first crystal structure, solved at 1.7 Å resolution, of the HprS cytoplasmic domains arranged as a homodimer. In both protomers, the catalytic ATP-binding domain contains a non-hydrolysable ATP analog coordinated by a magnesium ion. This structure allowed us to provide a detailed characterization of kinase-substrate interaction. Furthermore, the structural data are supported by biophysical studies of kinase interaction with cognate response regulator HprR and substrate ATP. The kinase activity is also assessed in the presence or absence of HprR.
Collapse
Affiliation(s)
- Anna Koczurowska
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 2/22, 90-537, Lodz, Poland
| | - David Ruiz Carrillo
- European Molecular Biology Laboratory, EMBL Hamburg, Notkestrasse 85, 22607, Hamburg, Germany
| | - María García Alai
- European Molecular Biology Laboratory, EMBL Hamburg, Notkestrasse 85, 22607, Hamburg, Germany
| | - Małgorzata Zakłos-Szyda
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 2/22, 90-537, Lodz, Poland
| | - Grzegorz Bujacz
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 2/22, 90-537, Lodz, Poland
| | - Agnieszka J Pietrzyk-Brzezinska
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 2/22, 90-537, Lodz, Poland.
| |
Collapse
|
20
|
Hrubý J, Dvořák Z. Binding of ligands to the aryl hydrocarbon receptor: An overview of methods. Toxicol Lett 2025; 404:37-46. [PMID: 39832617 DOI: 10.1016/j.toxlet.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/03/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor, which plays numerous and pivotal roles in human physiology and pathophysiology. Therefore, pharmacotherapeutic targeting of the AhR is a highly pertinent issue. The identification of new AhR ligands and the characterization of the interactions between the AhR ligands and AhR protein requires appropriate methodology. In spite the AhR is monomeric intracellular soluble receptor, the full-length human AhR protein has not been crystallized so far, and its isolation in a form applicable in the binding assays is highly challenging. Recent advances, including crystallization of AhR fragments, recombinant protein technologies, and cryogenic electron microscopy, allowed for exploitation of diverse experimental techniques for studying interactions between ligands and the AhR. In the current paper, we review existing AhR ligand binding assays, including their description, applicability and limitations.
Collapse
Affiliation(s)
- Jiří Hrubý
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc 783 71, Czech Republic
| | - Zdeněk Dvořák
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc 783 71, Czech Republic.
| |
Collapse
|
21
|
Mepperi J, Mukherjee S, Goel K, Kotamarthi HC. The Complex Energy Landscape of miRFP709, a 4 1-Knotted Protein, Results in Its Irreversible Denaturation. J Phys Chem B 2025; 129:1176-1184. [PMID: 39818792 DOI: 10.1021/acs.jpcb.4c05439] [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: 01/19/2025]
Abstract
Knotted proteins have a unique topological feature with an open knot, and the physiological significance of these knots remains elusive. In addition, these proteins challenge our understanding of the protein folding process, and whether they retain their native state during unfolding/refolding cycles like other proteins is debated. Most folding studies on knotted proteins have been performed on 31 and 52 knots, monitoring the tryptophan fluorescence. In this study, we probe the unfolding/refolding of a 41-knotted protein, miRFP709, which can be monitored through near-infrared fluorescence in addition to the intrinsic tryptophan emission. miRFP709, upon chemical unfolding and refolding, folds back to a compact, non-native, stable structure that loses its ability to bind to the biliverdin ligand and fluoresce. The refolded protein retains its secondary structure but behaves like a molten-globule state with an exposed hydrophobic surface. The complex folding landscape of these proteins results in hysteresis between the folding and refolding curves. We propose that upon refolding, either an altered knot or an unknotted structure prevents the formation of the native knotted structure.
Collapse
Affiliation(s)
- Jijith Mepperi
- Department of Chemistry, Indian Institute of Technology Madras, Chennai 600036, India
| | - Soham Mukherjee
- Department of Chemistry, Indian Institute of Technology Madras, Chennai 600036, India
| | - Khushboo Goel
- Department of Chemistry, Indian Institute of Technology Madras, Chennai 600036, India
| | | |
Collapse
|
22
|
Spiewla T, Grab K, Depaix A, Ziemkiewicz K, Warminski M, Jemielity J, Kowalska J. An MST-based assay reveals new binding preferences of IFIT1 for canonically and noncanonically capped RNAs. RNA (NEW YORK, N.Y.) 2025; 31:181-192. [PMID: 39643445 PMCID: PMC11789485 DOI: 10.1261/rna.080089.124] [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: 05/10/2024] [Accepted: 10/22/2024] [Indexed: 12/09/2024]
Abstract
IFITs (interferon-induced proteins with tetratricopeptide repeats) are components of the innate immune response that bind to viral and cellular RNA targets to inhibit translation and replication. The RNA target recognition is guided by molecular patterns, particularly at the RNA 5' ends. IFIT1 preferably binds RNAs modified with the m7G cap-0 structure, while RNAs with cap-1 structure are recognized with lower affinity. Less is known about the propensity of IFIT1 to recognize noncanonical RNA 5' ends, including hypermethylated and noncanonical RNA caps. Further insights into the structure-function relationship for IFIT1-RNA interactions are needed but require robust analytical methods. Here, we report a biophysical assay for quick, direct, in-solution affinity assessment of differently capped RNAs with IFIT1. The procedure, which relies on measuring microscale thermophoresis of fluorescently labeled protein as a function of increasing ligand concentration, is applicable to RNAs of various lengths and sequences without the need for their labeling or affinity tagging. Using the assay, we examined 13 canonically and noncanonically 5'-capped RNAs, revealing new binding preferences of IFIT1. The 5' terminal m6A mark in the m7G cap had a protective function against IFIT1, which was additive with the effect observed for the 2'-O position (m6Am cap-1). In contrast, an increased affinity for IFIT1 was observed for several noncanonical caps, including trimethylguanosine, unmethylated (G), and flavin-adenine dinucleotide caps. The results suggest new potential cellular targets of IFIT1 and may contribute to broadening the knowledge of the innate immune response mechanisms and the more effective design of chemically modified mRNAs.
Collapse
Affiliation(s)
- Tomasz Spiewla
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, 02-093 Warsaw, Poland
- Doctoral School of Exact and Natural Sciences, University of Warsaw, 02-089 Warsaw, Poland
| | - Katarzyna Grab
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, 02-093 Warsaw, Poland
- Doctoral School of Exact and Natural Sciences, University of Warsaw, 02-089 Warsaw, Poland
| | - Anais Depaix
- Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
| | - Kamil Ziemkiewicz
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, 02-093 Warsaw, Poland
- Doctoral School of Exact and Natural Sciences, University of Warsaw, 02-089 Warsaw, Poland
- Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
| | - Marcin Warminski
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, 02-093 Warsaw, Poland
| | - Jacek Jemielity
- Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
| | - Joanna Kowalska
- Division of Biophysics, Institute of Experimental Physics, Faculty of Physics, University of Warsaw, 02-093 Warsaw, Poland
| |
Collapse
|
23
|
Zuma LK, Pooe OJ, Mabaso NH, Alake J, Obakachi VA, Yakobi S, Gasa N, Karpoormath R, Simelane M. Assessing the efficacy of iso-mukaadial acetate and betulinic acid against selected Plasmodium falciparum glycolytic pathway proteins: in silico and in vitro studies. BMC Chem 2025; 19:16. [PMID: 39827161 PMCID: PMC11742793 DOI: 10.1186/s13065-025-01380-x] [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: 06/24/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025] Open
Abstract
Malaria is the extensive health concern in sub-Saharan Africa, with Plasmodium falciparum being the most lethal strain. The continued emergence of drug-resistant P. falciparum advocates for the development of new antimalarials. Our current study aimed to effectively explore the interaction capabilities of iso-mukaadial acetate (IMA) and betulinic acid (BA) against two essential P. falciparum glycolytic pathway proteins, PfLDH and PfHk. Recombinant PfLDH and PfHk were independently expressed in E. coli BL21 (DE3) cells and subsequently purified using affinity chromatography. Protein-ligand interaction studies probed in silico and in vitro approaches. Parasite inhibition studies confirmed potent antimalarial activity against the P. falciparum NF54 strains, with BA and IMA showing IC50 values of 1.27 µg/ml and 1.03 µg/ml against the asexual stage of P. falciparum, respectively. FTIR experiments confirmed interactions between the compounds and the secondary structure of the proteins. Direct protein-ligand interaction studies analysis using microscale thermophoresis (MST) showed a KD value of 0.1036 ± 0.6001 µM for the PfLDH-BA complex and 0.7473 ± 0.3554 µM KD value for PfLDH-IMA. Meanwhile, PfHk-IMA had 0.39701 ± 0.16298 µM KD value, while the PfHk-BA complex had no interaction detected. Molecular docking and molecular dynamics simulation studies were used to measure and confirm the interactive strength of complexes. Molecular docking reported a binding score of - 1.155 kcal/mol for the PfLDH-BA complex and a binding score of - 3.200 kcal/mol for PfLDH-IMA. The PfHk-BA complex had - 2.871 kcal/mol and PfHk-IMA complex had - 4.225 kcal/mol binding score. In conclusion, BA and IMA compounds had better interactions and remained bound within the binding sites of the glycolytic pathway proteins (PfLDH and PfHk).
Collapse
Affiliation(s)
- Lindiwe Khumbuzile Zuma
- Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Private Bag X54001, Durban, 4000, South Africa
| | - Ofentse Jacob Pooe
- Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Private Bag X54001, Durban, 4000, South Africa.
| | - Nonduduzo Hlengiwe Mabaso
- Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Private Bag X54001, Durban, 4000, South Africa
| | - John Alake
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Vincent A Obakachi
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Sinethemba Yakobi
- Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Private Bag X54001, Durban, 4000, South Africa
| | - Nothando Gasa
- Discipline of Biochemistry, School of Life Sciences, University of KwaZulu-Natal, Westville Campus, Private Bag X54001, Durban, 4000, South Africa
| | - Rajshekhar Karpoormath
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, 4000, South Africa
| | - Mthokozisi Simelane
- Department of Biochemistry, University of Johannesburg, Auckland Park Campus, Cnr Kingsway Avenue and University Road, Auckland, Park, PO Box 524, Johannesburg, 2006, South Africa
| |
Collapse
|
24
|
Trevisani M, Berselli A, Alberini G, Centonze E, Vercellino S, Cartocci V, Millo E, Ciobanu DZ, Braccia C, Armirotti A, Pisani F, Zara F, Castagnola V, Maragliano L, Benfenati F. A claudin5-binding peptide enhances the permeability of the blood-brain barrier in vitro. SCIENCE ADVANCES 2025; 11:eadq2616. [PMID: 39792664 PMCID: PMC11721574 DOI: 10.1126/sciadv.adq2616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 12/09/2024] [Indexed: 01/12/2025]
Abstract
The blood-brain barrier (BBB) maintains brain homeostasis but also prevents most drugs from entering the brain. No paracellular diffusion of solutes is allowed because of tight junctions that are made impermeable by the expression of claudin5 (CLDN5) by brain endothelial cells. The possibility of regulating the BBB permeability in a transient and reversible fashion is in strong demand for the pharmacological treatment of brain diseases. Here, we designed and tested short BBB-active peptides, derived from the CLDN5 extracellular domains and the CLDN5-binding domain of Clostridium perfringens enterotoxin, using a robust workflow of structural modeling and in vitro validation techniques. Computational analysis at the atom level based on solubility and affinity to CLDN5 identified a CLDN5-derived peptide not reported previously called f1-C5C2, which was soluble in biological media, displayed efficient binding to CLDN5, and transiently increased BBB permeability. The peptidomimetic strategy described here may have potential applications in the pharmacological treatment of brain diseases.
Collapse
Affiliation(s)
- Martina Trevisani
- Center for Synaptic Neuroscience and Technology (NSYN@UniGe), Istituto Italiano di Tecnologia, Largo Rosanna Benzi, 10, 16132 Genova, Italy
- Department of Experimental Medicine, Università degli Studi di Genova, Viale Benedetto XV, 3, 16132 Genova, Italy
| | - Alessandro Berselli
- Center for Synaptic Neuroscience and Technology (NSYN@UniGe), Istituto Italiano di Tecnologia, Largo Rosanna Benzi, 10, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132 Genova, Italy
| | - Giulio Alberini
- Center for Synaptic Neuroscience and Technology (NSYN@UniGe), Istituto Italiano di Tecnologia, Largo Rosanna Benzi, 10, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132 Genova, Italy
| | - Eleonora Centonze
- Center for Synaptic Neuroscience and Technology (NSYN@UniGe), Istituto Italiano di Tecnologia, Largo Rosanna Benzi, 10, 16132 Genova, Italy
| | - Silvia Vercellino
- Center for Synaptic Neuroscience and Technology (NSYN@UniGe), Istituto Italiano di Tecnologia, Largo Rosanna Benzi, 10, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132 Genova, Italy
| | - Veronica Cartocci
- Center for Synaptic Neuroscience and Technology (NSYN@UniGe), Istituto Italiano di Tecnologia, Largo Rosanna Benzi, 10, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132 Genova, Italy
| | - Enrico Millo
- Department of Experimental Medicine, Università degli Studi di Genova, Viale Benedetto XV, 3, 16132 Genova, Italy
| | - Dinu Zinovie Ciobanu
- Analytical Chemistry Facility, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Clarissa Braccia
- Analytical Chemistry Facility, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Andrea Armirotti
- Analytical Chemistry Facility, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Francesco Pisani
- Center for Synaptic Neuroscience and Technology (NSYN@UniGe), Istituto Italiano di Tecnologia, Largo Rosanna Benzi, 10, 16132 Genova, Italy
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari “Aldo Moro”, 70125 Bari, Italy
| | - Federico Zara
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genova, 16132 Genova, Italy
- Medical Genetics Unit, IRCCS Giannina Gaslini Institute, 16147 Genova, Italy
| | - Valentina Castagnola
- Center for Synaptic Neuroscience and Technology (NSYN@UniGe), Istituto Italiano di Tecnologia, Largo Rosanna Benzi, 10, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132 Genova, Italy
| | - Luca Maragliano
- Center for Synaptic Neuroscience and Technology (NSYN@UniGe), Istituto Italiano di Tecnologia, Largo Rosanna Benzi, 10, 16132 Genova, Italy
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Via Brecce Bianche, 60131 Ancona, Italy
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology (NSYN@UniGe), Istituto Italiano di Tecnologia, Largo Rosanna Benzi, 10, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132 Genova, Italy
| |
Collapse
|
25
|
Lindahl TL, Kumar AP, Hallström T, Al-Hashimi A, du Rietz A, Arlaman E, Uvdal K, Macwan AS. Dabigatran Attenuates the Binding of Thrombin to Platelets-A Novel Mechanism of Action. Thromb Haemost 2024. [PMID: 39586831 DOI: 10.1055/a-2483-0107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
BACKGROUND Thrombin is a multifunctional regulatory enzyme of the haemostasis and has both pro- and anticoagulant roles. It has, therefore, been a main target for drug discovery over many decades. Thrombin is a serine protease and possesses two positively charged regions called exosites, through which it is known to bind to many substrates. Dabigatran is a thrombin inhibitor and is widely used as an oral anticoagulant for the antithrombotic treatment of atrial fibrillation and venous thromboembolism. The mechanism by which dabigatran inhibits thrombin is the blockage of the active site, however, its effect on thrombin binding to its substrates has not been studied thoroughly and is thus poorly understood. MATERIAL AND METHODS The effect of dabigatran on thrombin binding to platelets was evaluated by flow cytometry using fluorescently labelled thrombin and washed platelets. Further, to confirm the results we utilized modern techniques for biomolecular binding studies, microscale thermophoresis (MST) and surface plasmon resonance (SPR), which validated the results. RESULTS Dabigatran inhibited thrombin binding to platelets as analysed by flow cytometry. The inhibition was dose dependent with IC50 of 118 nM which was slightly lower than for inhibition of platelet activation and is close to the clinically relevant plasma concentration of dabigatran. MST and SPR also confirmed inhibitory effect of dabigatran on thrombin binding to platelets. CONCLUSION Apart from blocking the active site, dabigatran also inhibits thrombin binding to platelets. Since thrombin has numerous functions beyond the cardiovascular system, this finding may have important implications.
Collapse
Affiliation(s)
- Tomas L Lindahl
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Aishwarya Prasanna Kumar
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | | | - Ahmed Al-Hashimi
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Anna du Rietz
- Division of Molecular Surface Physics and Nanoscience, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Elena Arlaman
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Kajsa Uvdal
- Division of Molecular Surface Physics and Nanoscience, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Ankit S Macwan
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
26
|
Zhang Z, Luo H, Zhang X, Yang R, Yan S, Yang Q, Yang J. Extracellular Vesicles Mimetic Design of Membrane Chimeric Nanovesicles for dsRNA Delivery in Spray-Induced Gene Silencing for Crop Protection. ACS NANO 2024; 18:32468-32480. [PMID: 39530910 DOI: 10.1021/acsnano.4c06282] [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: 11/16/2024]
Abstract
Spray-induced gene silencing (SIGS) presents a promising RNA interference (RNAi)-based crop protection strategy against eukaryotic phytopathogens. However, the application of SIGS faces challenges, such as the limited uptake of dsRNA by certain pathogens and the instability of dsRNA in the environment. This study introduces innovative biomimetic nanovesicles, called extracellular vesicle (EV) mimetic chimeric nanovesicles (ECNs), assembled from tomato leaf cell membranes and cationic sterosomes via the freeze-thaw method. Similar to the function of EVs in nucleic acid transport between cells, ECNs serve as a hybrid nanosystem to overcome the challenge of delivering exogenous dsRNA in Phytophthora infestans. When applied to SIGS, the superiority of ECNs in crop protection becomes more apparent, including high loading and protection of dsRNA, improved biosafety, and efficient internalization into pathogen and plant cells, all of which significantly enhance the efficacy of RNAi in preventing early infection of P. infestans to susceptible tomato plants. This study demonstrates that ECNs are promising RNA delivery vehicles and will promote the use of SIGS-based RNA pesticides in sustainable agricultural production.
Collapse
Affiliation(s)
- Zheng Zhang
- MOE Key Laboratory of Intelligent Biomanufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, P.R. China
| | - Hongye Luo
- MOE Key Laboratory of Intelligent Biomanufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, P.R. China
| | - Xinyuan Zhang
- MOE Key Laboratory of Intelligent Biomanufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, P.R. China
| | - Run Yang
- MOE Key Laboratory of Intelligent Biomanufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, P.R. China
| | - Shili Yan
- MOE Key Laboratory of Intelligent Biomanufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, P.R. China
| | - Qing Yang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518120, P.R. China
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing 100193, P.R. China
| | - Jun Yang
- MOE Key Laboratory of Intelligent Biomanufacturing, School of Bioengineering, Dalian University of Technology, Dalian 116024, P.R. China
| |
Collapse
|
27
|
Tang X, Chen M, Li X, Lu H, Zhang X, Li Y, Li J, Xiao Y, Qin Z. A Mechanistic Approach on Perception Mode of ABA Receptors (PYLs) to Novel Opabactin Analogues. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024. [PMID: 39563176 DOI: 10.1021/acs.jafc.4c07265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
This study explored the structural mechanisms governing the binding of opabactin (OP) analogues 2-6 to abscisic acid (ABA) receptors by employing a combination of micro-scale thermophoresis (MST), phosphatase activity inhibition assays, and molecular dynamics simulations. The compounds 3-6 selectively activated PYR1, PYL2, and PYL6, while exhibiting minimal activity against PYL10, thus identifying them as selective ABA receptor agonists. Additionally, these analogues exerted a significant inhibitory effect on the phosphatase HAB1 upon binding to the receptors. The molecular dynamics simulations further elucidated the detailed binding interactions between various OP analogues and the ABA receptor PYR1, highlighting their role in inducing conformational changes within the receptor. Specifically, the study focused on the facilitation of the closure of the Gate and CL1 loops and the fine-tuning of the Latch loop to enhance the plasticity of the binding pocket, thereby influencing receptor-ligand interactions. The investigation emphasized the critical role of conserved water molecules in stabilizing the ligand-PYLs-PP2Cs complexes. Furthermore, free energy decomposition calculations demonstrated that the ligand's affinity was significantly affected by its ability to establish polar contacts between the polar groups within the ligand tail and the residues at the base of the binding pocket. This research lays a robust foundation for the development of novel ABA functional analogues with improved activity.
Collapse
Affiliation(s)
- Xianjun Tang
- College of Science, China Agricultural University, Beijing 100193, China
| | - Minghui Chen
- College of Science, China Agricultural University, Beijing 100193, China
| | - Xiaobin Li
- College of Science, China Agricultural University, Beijing 100193, China
| | - Huizhe Lu
- College of Science, China Agricultural University, Beijing 100193, China
| | - Xueqin Zhang
- College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Yiyi Li
- College of Science, China Agricultural University, Beijing 100193, China
| | - Jiaqi Li
- College of Science, China Agricultural University, Beijing 100193, China
| | - Yumei Xiao
- College of Science, China Agricultural University, Beijing 100193, China
| | - Zhaohai Qin
- College of Science, China Agricultural University, Beijing 100193, China
| |
Collapse
|
28
|
Rahman A, Tamseel S, Dutta S, Khan N, Faaiz M, Rastogi H, Nath JR, Haldar K, Chowdhury P, Ashish, Bhattacharjee S. Artemisinin-resistant Plasmodium falciparum Kelch13 mutant proteins display reduced heme-binding affinity and decreased artemisinin activation. Commun Biol 2024; 7:1499. [PMID: 39538019 PMCID: PMC11561146 DOI: 10.1038/s42003-024-07178-2] [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: 02/19/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
The potency of frontline antimalarial drug artemisinin (ART) derivatives is triggered by heme-induced cleavage of the endoperoxide bond to form reactive heme-ART alkoxy radicals and covalent heme-ART adducts, which are highly toxic to the parasite. ART-resistant (ART-R) parasites with mutations in the Plasmodium falciparum Kelch-containing protein Kelch13 (PfKekch13) exhibit impaired hemoglobin uptake, reduced yield of hemoglobin-derived heme, and thus decreased ART activation. However, any direct involvement of PfKelch13 in heme-mediated ART activation has not been reported. Here, we show that the purified recombinant PfKelch13 wild-type (WT) protein displays measurable binding affinity for iron and heme, the main effectors for ART activation. The heme-binding property is also exhibited by the native PfKelch13 protein from parasite culture. The two ART-R recombinant PfKelch13 mutants (C580Y and R539T) display weaker heme binding affinities compared to the ART-sensitive WT and A578S mutant proteins, which further translates into reduced yield of heme-ART derivatives when ART is incubated with the heme molecules bound to the mutant PfKelch13 proteins. In conclusion, this study provides the first evidence for ART activation via the heme-binding propensity of PfKelch13. This mechanism may contribute to the modulation of ART-R levels in malaria parasites through a novel function of PfKelch13.
Collapse
Affiliation(s)
- Abdur Rahman
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, Delhi, 110067, India
| | - Sabahat Tamseel
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, Delhi, 110067, India
| | - Smritikana Dutta
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, Delhi, 110067, India
| | - Nawaal Khan
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, Delhi, 110067, India
| | - Mohammad Faaiz
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, Delhi, 110067, India
| | - Harshita Rastogi
- Department of Chemistry, Indian Institute of Technology (IIT) Delhi, Hauz Khas, New Delhi, 110016, India
| | - Jyoti Rani Nath
- Department of Chemistry, Indian Institute of Technology (IIT) Delhi, Hauz Khas, New Delhi, 110016, India
| | - Kasturi Haldar
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, IN, USA
| | - Pramit Chowdhury
- Department of Chemistry, Indian Institute of Technology (IIT) Delhi, Hauz Khas, New Delhi, 110016, India
| | - Ashish
- Council of Scientific and Industrial Research-Institute of Microbial Technology, Chandigarh, 160036, India
| | - Souvik Bhattacharjee
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, Delhi, 110067, India.
| |
Collapse
|
29
|
Stangherlin S, Ding Y, Liu J. Dissociation Constant (K d) Measurement for Small-Molecule Binding Aptamers: Homogeneous Assay Methods and Critical Evaluations. SMALL METHODS 2024:e2401572. [PMID: 39511863 DOI: 10.1002/smtd.202401572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/27/2024] [Indexed: 11/15/2024]
Abstract
Since 1990, numerous aptamers have been isolated and discovered for use in various analytical, biomedical, and environmental applications. This trend continues to date. A critical step in the characterization of aptamer binding is to measure its binding affinity toward both target and non-target molecules. Dissociation constant (Kd) is the most commonly used value in characterizing aptamer binding. In this article, homogenous assays are reviewed for aptamers that can bind small-molecule targets. The reviewed methods include label-free methods, such as isothermal titration calorimetry, intrinsic fluorescence of target molecules, DNA staining dyes, and nuclease digestion assays, and labeled methods, such as the strand displacement reaction. Some methods are not recommended, such as those based on the aggregation of gold nanoparticles and the desorption of fluorophore-labeled DNA from nanomaterials. The difference between the measured apparent Kd and the true Kd of aptamer binding is stressed. In addition, avoiding the titration regime and paying attention to the time required to reach equilibrium are discussed. Finally, it is important to include mutated non-binding sequences as controls.
Collapse
Affiliation(s)
- Stefen Stangherlin
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Yuzhe Ding
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Juewen Liu
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| |
Collapse
|
30
|
Bonomini A, Felicetti T, Pacetti M, Bertagnin C, Coletti A, Giammarino F, De Angelis M, Poggialini F, Macchiarulo A, Sabatini S, Mercorelli B, Nencioni L, Vicenti I, Dreassi E, Cecchetti V, Tabarrini O, Loregian A, Massari S. Optimization of potent, broad-spectrum, and specific anti-influenza compounds targeting RNA polymerase PA-PB1 heterodimerization. Eur J Med Chem 2024; 277:116737. [PMID: 39153334 DOI: 10.1016/j.ejmech.2024.116737] [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: 06/24/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/19/2024]
Abstract
Influenza viruses (IV) are single-stranded RNA viruses with a negative-sense genome and have the potential to cause pandemics. While vaccines exist for influenza, their protection is only partial. Additionally, there is only a limited number of approved anti-IV drugs, which are associated to emergence of drug resistance. To address these issues, for years we have focused on the development of small-molecules that can interfere with the heterodimerization of PA and PB1 subunits of the IV RNA-dependent RNA polymerase (RdRP). In this study, starting from a cycloheptathiophene-3-carboxamide compound that we recently identified, we performed iterative cycles of medicinal chemistry optimization that led to the identification of compounds 43 and 45 with activity in the nanomolar range against circulating A and B strains of IV. Mechanistic studies demonstrated the ability of 43 and 45 to interfere with viral RdRP activity by disrupting PA-PB1 subunits heterodimerization and to bind to the PA C-terminal domain through biophysical assays. Most important, ADME studies of 45 also showed an improvement in the pharmacokinetic profile with respect to the starting hit.
Collapse
Affiliation(s)
- Anna Bonomini
- Department of Molecular Medicine, University of Padua, 35121, Padua, Italy
| | - Tommaso Felicetti
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Martina Pacetti
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Chiara Bertagnin
- Department of Molecular Medicine, University of Padua, 35121, Padua, Italy
| | - Alice Coletti
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Federica Giammarino
- Department of Medical Biotechnologies, University of Siena, 53100, Siena, Italy
| | - Marta De Angelis
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185, Rome, Italy
| | - Federica Poggialini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100, Siena, Italy
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Stefano Sabatini
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | | | - Lucia Nencioni
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185, Rome, Italy
| | - Ilaria Vicenti
- Department of Medical Biotechnologies, University of Siena, 53100, Siena, Italy
| | - Elena Dreassi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100, Siena, Italy
| | - Violetta Cecchetti
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Oriana Tabarrini
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, 35121, Padua, Italy.
| | - Serena Massari
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy.
| |
Collapse
|
31
|
El Atab O, Gupta B, Han Z, Stribny J, Asojo OA, Schneiter R. Alpha-1-B glycoprotein (A1BG) inhibits sterol-binding and export by CRISP2. J Biol Chem 2024; 300:107910. [PMID: 39433128 PMCID: PMC11599453 DOI: 10.1016/j.jbc.2024.107910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 10/04/2024] [Accepted: 10/14/2024] [Indexed: 10/23/2024] Open
Abstract
Proteins belonging to the CAP superfamily are present in all kingdoms of life and have been implicated in various processes, including sperm maturation and cancer progression. They are mostly secreted glycoproteins and share a unique conserved CAP domain. The precise mode of action of these proteins, however, has remained elusive. Saccharomyces cerevisiae expresses three members of this protein family, which bind sterols in vitro and promote sterol secretion from cells. This sterol-binding and export function of yeast Pry proteins is conserved in the mammalian cysteine-rich secretory protein (CRISP) proteins and other CAP superfamily members. CRISP3 is an abundant protein of the human seminal plasma and interacts with alpha-1-B glycoprotein (A1BG), a human plasma glycoprotein that is upregulated in different types of cancers. Here, we examined whether the interaction between CRISP proteins and A1BG affects the sterol-binding function of CAP family members. Coexpression of A1BG with CAP proteins abolished their sterol export function in yeast and their interaction inhibits sterol-binding in vitro. We map the interaction between A1BG and CRISP2 to the third of five repeated immunoglobulin-like domains within A1BG. Interestingly, the interaction between A1BG and CRISP2 requires magnesium, suggesting that coordination of Mg2+ by the highly conserved tetrad residues within the CAP domain is essential for a stable interaction between the two proteins. The observation that A1BG modulates the sterol-binding function of CRISP2 has potential implications for the role of A1BG and related immunoglobulin-like domain containing proteins in cancer progression and the toxicity of reptile venoms containing CRISP proteins.
Collapse
Affiliation(s)
- Ola El Atab
- Department of Biology, University of Fribourg, Chemin du Musée 10, Fribourg, Switzerland
| | - Barkha Gupta
- Department of Biology, University of Fribourg, Chemin du Musée 10, Fribourg, Switzerland
| | - Zhu Han
- Department of Biology, University of Fribourg, Chemin du Musée 10, Fribourg, Switzerland
| | - Jiri Stribny
- Department of Biology, University of Fribourg, Chemin du Musée 10, Fribourg, Switzerland
| | | | - Roger Schneiter
- Department of Biology, University of Fribourg, Chemin du Musée 10, Fribourg, Switzerland.
| |
Collapse
|
32
|
Choi ES, Hnath B, Sha CM, Dokholyan NV. Unveiling the double-edged sword: SOD1 trimers possess tissue-selective toxicity and bind septin-7 in motor neuron-like cells. Structure 2024; 32:1776-1792.e5. [PMID: 39208794 PMCID: PMC11455619 DOI: 10.1016/j.str.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/10/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Misfolded species of superoxide dismutase 1 (SOD1) are associated with increased death in amyotrophic lateral sclerosis (ALS) models compared to insoluble protein aggregates. The mechanism by which structurally independent SOD1 trimers cause cellular toxicity is unknown but may drive disease pathology. Here, we uncovered the SOD1 trimer interactome-a map of potential tissue-selective protein-binding partners in the brain, spinal cord, and skeletal muscle. We identified binding partners and key pathways associated with SOD1 trimers and found that trimers may affect normal cellular functions such as dendritic spine morphogenesis and synaptic function in the central nervous system and cellular metabolism in skeletal muscle. We discovered SOD1 trimer-selective enrichment of genes. We performed detailed computational and biochemical characterization of SOD1 trimer protein binding for septin-7. Our investigation highlights key proteins and pathways within distinct tissues, revealing a plausible intersection of genetic and pathophysiological mechanisms in ALS through interactions involving SOD1 trimers.
Collapse
Affiliation(s)
- Esther Sue Choi
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA; Medical Scientist Training Program, Penn State College of Medicine, Hershey, PA, USA
| | - Brianna Hnath
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA; Department of Biomedical Engineering, Penn State University, University Park, PA, USA
| | - Congzhou Mike Sha
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA; Medical Scientist Training Program, Penn State College of Medicine, Hershey, PA, USA
| | - Nikolay V Dokholyan
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA; Department of Biomedical Engineering, Penn State University, University Park, PA, USA; Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA; Department of Chemistry, Penn State University, University Park, PA, USA.
| |
Collapse
|
33
|
Agarwal A, Muniyappa K. Mycobacterium smegmatis putative Holliday junction resolvases RuvC and RuvX play complementary roles in the processing of branched DNA structures. J Biol Chem 2024; 300:107732. [PMID: 39222685 PMCID: PMC11466669 DOI: 10.1016/j.jbc.2024.107732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/14/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024] Open
Abstract
In eubacteria, Holliday junction (HJ) resolvases (HJRs) are crucial for faithful segregation of newly replicated chromosomes, homologous recombination, and repair of stalled/collapsed DNA replication forks. However, compared with the Escherichia coli HJRs, little is known about their orthologs in mycobacterial species. A genome-wide analysis of Mycobacterium smegmatis identified two genes encoding putative HJRs, namely RuvC (MsRuvC) and RuvX (MsRuvX); but whether they play redundant, overlapping, or distinct roles remains unknown. Here, we reveal that MsRuvC exists as a homodimer while MsRuvX as a monomer in solution, and both showed high-binding affinity for branched DNAs compared with unbranched DNA species. Interestingly, the DNA cleavage specificities of MsRuvC and MsRuvX were found to be mutually exclusive: the former efficiently promotes HJ resolution, in a manner analogous to the Escherichia coli RuvC, but does not cleave other branched DNA species; whereas the latter is a versatile DNase capable of cleaving a variety of branched DNA structures, including 3' and 5' flap DNA, splayed-arm DNA and dsDNA with 3' and 5' overhangs but lacks the HJ resolution activity. Point mutations in the RNase H-like domains of MsRuvC and MsRuvX pinpointed critical residues required for their DNA cleavage activities and also demonstrated uncoupling between DNA-binding and DNA cleavage activities. Unexpectedly, we found robust evidence that MsRuvX possesses a double-strand/single-strand junction-specific endonuclease and ssDNA exonucleolytic activities. Combined, our findings highlight that the RuvC and RuvX DNases play distinct complementary, and not redundant, roles in the processing of branched DNA structures in M. smegmatis.
Collapse
Affiliation(s)
- Ankit Agarwal
- Department of Biochemistry, Indian Institute of Science, Bengaluru, India
| | - Kalappa Muniyappa
- Department of Biochemistry, Indian Institute of Science, Bengaluru, India.
| |
Collapse
|
34
|
Zhang T, Liu Y, Cao J, Jiang L, Wang P, Ren F, Yi H. Exploration of dynamic interaction between β-lactoglobulin and casein micelles during UHT milk process. Int J Biol Macromol 2024; 277:134367. [PMID: 39089562 DOI: 10.1016/j.ijbiomac.2024.134367] [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: 04/19/2024] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
The protein aggregation induced by UHT treatment shortens the shelf life of UHT milk. However, the mechanism of β-Lg induced casein micelle aggregation remains unclear. Herein, the dynamic interaction between β-Lg and casein micelles during UHT processing was investigated by experimental techniques and molecular dynamics simulations. Results showed that β-Lg decreased the stability of casein micelles, increased their size and zeta potential. Raman and FTIR spectra analysis suggested that hydrogen and disulfide bonds facilitated their interaction. Cryo-TEM showed that the formation of the casein micelle/β-Lg complex involved rigid binding, flexible linking, and severe cross-linking aggregation during UHT processing. SAXS and MST demonstrated β-Lg bound to κ-casein on micelle surfaces with a dissociation constant (Kd) of 3.84 ± 1.14 μm. Molecular docking and dynamic simulations identified the interacting amino acid residues and clarified that electrostatic and van der Waals forces drove the interaction. UHT treatment increased hydrogen bonds and decreased total binding energy. The non-covalent binding promoted the formation of disulfide bonds between β-Lg and casein micelles under heat treatment. Ultimately, it was concluded that non-covalent interaction and disulfide bonding resulted in casein micelle/β-Lg aggregates. These findings provided scientific insights into protein aggregation in UHT milk.
Collapse
Affiliation(s)
- Tai Zhang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong Province, China; Food Laboratory of Zhongyuan, Luohe 462300, Henan, China
| | - Yisuo Liu
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong Province, China; Food Laboratory of Zhongyuan, Luohe 462300, Henan, China
| | - Jiayuan Cao
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong Province, China
| | - Lu Jiang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong Province, China
| | - Pengjie Wang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, China; Food Laboratory of Zhongyuan, Luohe 462300, Henan, China
| | - Fazheng Ren
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, China; Food Laboratory of Zhongyuan, Luohe 462300, Henan, China.
| | - Huaxi Yi
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao 266003, Shandong Province, China; Food Laboratory of Zhongyuan, Luohe 462300, Henan, China.
| |
Collapse
|
35
|
Collins M, Ibeanu N, Grabowska WR, Awwad S, Khaw PT, Brocchini S, Khalili H. Bispecific FpFs: a versatile tool for preclinical antibody development. RSC Chem Biol 2024:d4cb00130c. [PMID: 39347456 PMCID: PMC11427889 DOI: 10.1039/d4cb00130c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/08/2024] [Indexed: 10/01/2024] Open
Abstract
We previously described FpFs 1̲ (Fab-PEG-Fab) as binding mimetics of IgGs. FpFs are prepared with di(bis-sulfone) conjugation reagents 3̲ that undergo disulfide rebridging conjugation with the accessible disulfide of each Fab (Scheme 1). We have now prepared bispecific FpFs 2̲ (bsFpF and Fab1-PEG-Fab2) as potential bispecific antibody mimetics with the intent that bsFpFs could be used in preclinical antibody development since sourcing bispecific antibodies may be challenging during preclinical research. The di(bis-sulfone) reagent 3̲ was first used to prepare a bsFpF 2̲ by the sequential conjugation of a first Fab and then a second Fab to another target (Scheme 2). Seeking to improve bsFpF synthesis, the asymmetric conjugation reagent, bis-sulfone bis-sulfide 1̲6̲, with different thiol conjugation reactivities at each terminus (Scheme 4) was examined and the bsFpFs appeared to be formed at similar conversion to the di(bis-sulfone) reagent 3̲. To explore the advantages of using common intermediates in the preparation of bsFpF families, we investigated bsFpF synthesis with a protein conjugation-ligation approach (Scheme 5). Reagents with a bis-sulfone moiety for conjugation on one PEG terminus and a ligation moiety on the other terminus were examined. Bis-sulfone PEG trans-cyclooctene (TCO) 2̲8̲ and bis-sulfone PEG tetrazine (Tz) 3̲0̲ were used to prepare several bsFpFs targeting various therapeutic targets (TNF-α, IL6R, IL17, and VEGF) and tissue affinity targets (hyaluronic acid and collagen II). Surface plasmon resonance (SPR) binding studies indicated that there was little difference between the dissociation rate constant (k d) for the unmodified Fab, mono-conjugated PEG-Fab and the corresponding Fab in a bsFpF. The Fab association rate (k a) in the bsFpF was slower than for PEG-Fab, which may be because of mass differences that influence SPR results. These observations suggest that each Fab will bind to its target independently of the other Fab and that bsFpF binding profiles can be estimated using the corresponding PEG-Fab conjugates.
Collapse
Affiliation(s)
- Matthew Collins
- School of Health, Sport and Bioscience, University of East London London UK
| | - Nkiru Ibeanu
- School of Pharmacy, University College London London UK
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology London EC1V 9EL UK
| | | | - Sahar Awwad
- School of Pharmacy, University College London London UK
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology London EC1V 9EL UK
| | - Peng T Khaw
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology London EC1V 9EL UK
| | | | - Hanieh Khalili
- School of Pharmacy, University College London London UK
- School of Biomedical Science, University of West London London W5 5RF UK
| |
Collapse
|
36
|
Dodd RJ, Blundell CD, Sattelle BM, Enghild JJ, Milner CM, Day AJ. Chemical modification of hyaluronan oligosaccharides differentially modulates hyaluronan-hyaladherin interactions. J Biol Chem 2024; 300:107668. [PMID: 39128716 PMCID: PMC11460632 DOI: 10.1016/j.jbc.2024.107668] [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: 03/17/2024] [Revised: 08/04/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024] Open
Abstract
The glycosaminoglycan hyaluronan (HA) is a ubiquitous, nonsulfated polysaccharide with diverse biological roles mediated through its interactions with HA-binding proteins (HABPs). Most HABPs belong to the Link module superfamily, including the major HA receptor, CD44, and secreted protein TSG-6, which catalyzes the covalent transfer of heavy chains from inter-α-inhibitor onto HA. The structures of the HA-binding domains (HABDs) of CD44 (HABD_CD44) and TSG-6 (Link_TSG6) have been determined and their interactions with HA extensively characterized. The mechanisms of binding are different, with Link_TSG6 interacting with HA primarily via ionic and CH-π interactions, whereas HABD_CD44 binds solely via hydrogen bonds and van der Waals forces. Here, we exploit these differences to generate HA oligosaccharides, chemically modified at their reducing ends, that bind specifically and differentially to these target HABPs. Hexasaccharides (HA6AN) modified with 2- or 3-aminobenzoic acid (HA6-2AA, HA6-3AA) or 2-amino-4-methoxybenzoic acid (HA6-2A4MBA), had increased affinities for Link_TSG6 compared to unmodified HA6AN. These modifications did not increase the affinity for CD44_HABD. A model of HA6-2AA (derived from the solution dynamic 3D structure of HA4-2AA) was docked into the Link_TSG6 structure, providing evidence that the 2AA-carboxyl forms a salt bridge with Arginine-81. These modeling results informed a second series of chemical modifications for HA oligosaccharides, which again showed differential binding to the two proteins. Several modifications to HA4 and HA6 were found to convert the oligosaccharide into substrates for heavy chain transfer, whereas unmodified HA4 and HA6 are not. This study has generated valuable research tools to further understand HA biology.
Collapse
Affiliation(s)
- Rebecca J Dodd
- Wellcome Centre for Cell Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom; Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | | | | | - Jan J Enghild
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Caroline M Milner
- Wellcome Centre for Cell Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom; Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom; Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom
| | - Anthony J Day
- Wellcome Centre for Cell Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom; Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom; Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
37
|
Saade C, Pozza A, Bonneté F, Finet S, Lutz-Bueno V, Tully MD, Varela PF, Lacapère JJ, Combet S. Enhanced structure/function of mTSPO translocator in lipid:surfactant mixed micelles. Biochimie 2024; 224:3-15. [PMID: 38663457 DOI: 10.1016/j.biochi.2024.04.008] [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: 02/05/2024] [Revised: 04/06/2024] [Accepted: 04/19/2024] [Indexed: 05/06/2024]
Abstract
TSPO is a ubiquitous transmembrane protein used as a pharmacological marker in neuroimaging. The only known atomic structure of mammalian TSPOs comes from the solution NMR of mouse TSPO (mTSPO) bound to the PK11195 ligand and in a DPC surfactant environment. No structure is available in a biomimetic environment and without PK11195 which strongly stiffens the protein. We measured the effect of different amphiphilic environments on ligand-free mTSPO to study its structure/function and find optimal solubilization conditions. By replacing the SDS surfactant, where the recombinant protein is purified, with mixed lipid:surfactant (DMPC:DPC) micelles at different ratios (0:1, 1:2, and 2:1, w:w), the α-helix content and interactions and the intrinsic tryptophan (Trp) fluorescence of mTSPO are gradually increased. Small-angle X-ray scattering (SAXS) shows a more extended mTSPO/belt complex with the addition of lipids: Dmax ∼95 Å in DPC alone versus ∼142 Å in DMPC:DPC (1:2). SEC-MALLS shows that the molecular composition of the mTSPO belt is ∼98 molecules for DPC alone and ∼58 DMPC and ∼175 DPC for DMPC:DPC (1:2). Additionally, DMPC:DPC micelles stabilize mTSPO compared to DPC alone, where the protein has a greater propensity to aggregate. These structural changes are consistent with the increased affinity of mTSPO for the PK11195 ligand in presence of lipids (Kd ∼70 μM in DPC alone versus ∼0.91 μM in DMPC:DPC, 1:2), as measured by microscale thermophoresis (MST). In conclusion, mixed lipid:surfactant micelles open new possibilities for the stabilization of membrane proteins and for their study in solution in a more biomimetic amphiphilic environment.
Collapse
Affiliation(s)
- Christelle Saade
- Laboratoire Léon-Brillouin (LLB), UMR12 CEA-CNRS, Université Paris-Saclay, F-91191, Gif-sur-Yvette CEDEX, France
| | - Alexandre Pozza
- Université Paris Cité, CNRS UMR7099, Biochimie des Protéines Membranaires, F-75005, Paris, France
| | - Françoise Bonneté
- Université Paris Cité, CNRS UMR7099, Biochimie des Protéines Membranaires, F-75005, Paris, France
| | - Stéphanie Finet
- Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie (IMPMC), UMR 7590 CNRS-Sorbonne Université Université, MNHN, IRD, F-75005, Paris, France
| | - Viviane Lutz-Bueno
- Laboratoire Léon-Brillouin (LLB), UMR12 CEA-CNRS, Université Paris-Saclay, F-91191, Gif-sur-Yvette CEDEX, France; Paul Scherrer Institut (PSI), Forschungsstrasse 111, 5232, Villigen PSI, Switzerland
| | - Mark D Tully
- The European Synchrotron (ESRF), 71 Avenue des Martyrs, F-38043, Grenoble, France
| | - Paloma F Varela
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, F-91191, Gif-sur-Yvette CEDEX, France
| | - Jean-Jacques Lacapère
- Sorbonne Université, Ecole Normale Supérieure, PSL University, CNRS UMR 7203, Laboratoire des BioMolécules (LBM), 4 Place Jussieu, F-75005, Paris, France
| | - Sophie Combet
- Laboratoire Léon-Brillouin (LLB), UMR12 CEA-CNRS, Université Paris-Saclay, F-91191, Gif-sur-Yvette CEDEX, France.
| |
Collapse
|
38
|
Wang K, Zhang L, Zhang S, Liu Y, Mao J, Liu Z, Xu L, Li K, Wang J, Ma Y, Wang J, Li H, Wang Z, Li G, Cheng H, Ye M. Metabolic labeling based methylome profiling enables functional dissection of histidine methylation in C3H1 zinc fingers. Nat Commun 2024; 15:7459. [PMID: 39198440 PMCID: PMC11358137 DOI: 10.1038/s41467-024-51979-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024] Open
Abstract
Protein methylation is a functionally important post-translational modification that occurs on diverse amino acid residues. The current proteomics approaches are inefficient to discover the methylation on residues other than Arg and Lys, which hinders the deep understanding of the functional role of rare protein methylation. Herein, we present a methyl-specific metabolic labeling approach for global methylome mapping, which enable the acquisition of methylome dataset covering diverse methylation types. Interestingly, of the identified methylation events, His methylation is found to be preferably occurred in C3H1 zinc fingers (ZFs). These His methylation events are determined to be Nπ specific and catalyzed by CARNMT1. The His methylation is found to stabilize the structure of ZFs. U2AF1 is used as a proof-of-concept to highlight the functional importance of His methylation in ZFs in RNA binding and RNA metabolism. The results of this study enable novel understanding of how protein methylation regulates cellular processes.
Collapse
Grants
- This work was supported, in part, by funds from the China State Key Basic Research Program Grants (2021YFA13026012, 2019YFA0709400, 2022YFA1303300), the Strategic Priority Research Program of Chinese Academy of Sciences (XDB37040401, XDB0570100), the National Natural Science Foundation of China (21804131, 92153302, 21933010, 31925008), the innovation program (DICP I202226) of science and research from the DICP, CAS.
Collapse
Affiliation(s)
- Keyun Wang
- State Key Laboratory of Medical Proteomics, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Li Zhang
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Sirui Zhang
- CAS Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ye Liu
- Laboratory of Molecular Modeling and Design, State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Jiawei Mao
- State Key Laboratory of Medical Proteomics, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhen Liu
- State Key Laboratory of Medical Proteomics, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lin Xu
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Kejia Li
- State Key Laboratory of Medical Proteomics, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jianshu Wang
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yanni Ma
- State Key Laboratory of Medical Proteomics, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiayi Wang
- State Key Laboratory of Medical Proteomics, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haitao Li
- MOE Key Laboratory of Protein Sciences, Beijing Frontier Research Center for Biological Structure, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Zefeng Wang
- CAS Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
- School of Life Science, Southern University of Science and Technology, Shenzhen, China.
| | - Guohui Li
- Laboratory of Molecular Modeling and Design, State Key Laboratory of Molecular Reaction Dynamics, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.
| | - Hong Cheng
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| | - Mingliang Ye
- State Key Laboratory of Medical Proteomics, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
39
|
Alieva RT, Ulasov AV, Khramtsov YV, Slastnikova TA, Lupanova TN, Gribova MA, Georgiev GP, Rosenkranz AA. Optimization of a Modular Nanotransporter Design for Targeted Intracellular Delivery of Photosensitizer. Pharmaceutics 2024; 16:1083. [PMID: 39204428 PMCID: PMC11360004 DOI: 10.3390/pharmaceutics16081083] [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: 06/30/2024] [Revised: 07/26/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
Modular nanotransporters (MNTs) are drug delivery systems for targeted cancer treatment. As MNTs are composed of several modules, they offer the advantage of high specificity and biocompatibility in delivering drugs to the target compartment of cancer cells. The large carrier module brings together functioning MNT modules and serves as a platform for drug attachment. The development of smaller-sized MNTs via truncation of the carrier module appears advantageous in facilitating tissue penetration. In this study, two new MNTs with a truncated carrier module containing either an N-terminal (MNTN) or a C-terminal (MNTC) part were developed by genetic engineering. Both new MNTs demonstrated a high affinity for target receptors, as revealed by fluorescent-labeled ligand-competitive binding. The liposome leakage assay proved the endosomolytic activity of MNTs. Binding to the importin heterodimer of each truncated MNT was revealed by a thermophoresis assay, while only MNTN possessed binding to Keap1. Finally, the photodynamic efficacy of the photosensitizer attached to MNTN was significantly higher than when attached to either MNTC or the original MNTs. Thus, this work reveals that MNT's carrier module can be truncated without losing MNT functionality, favoring the N-terminal part of the carrier module due to its ability to bind Keap1.
Collapse
Affiliation(s)
- Rena T. Alieva
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology of Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia
| | - Alexey V. Ulasov
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology of Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia
| | - Yuri V. Khramtsov
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology of Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia
| | - Tatiana A. Slastnikova
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology of Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia
| | - Tatiana N. Lupanova
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology of Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia
| | - Maria A. Gribova
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology of Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory St., 119234 Moscow, Russia
| | - Georgii P. Georgiev
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology of Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia
| | - Andrey A. Rosenkranz
- Laboratory of Molecular Genetics of Intracellular Transport, Institute of Gene Biology of Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, 1-12 Leninskie Gory St., 119234 Moscow, Russia
| |
Collapse
|
40
|
Huang P, Ji F, Cheung AHK, Fu K, Zhou Q, Ding X, Chen D, Lin Y, Wang L, Jiao Y, Chu ESH, Kang W, To KF, Yu J, Wong CC. Peptostreptococcus stomatis promotes colonic tumorigenesis and receptor tyrosine kinase inhibitor resistance by activating ERBB2-MAPK. Cell Host Microbe 2024; 32:1365-1379.e10. [PMID: 39059397 DOI: 10.1016/j.chom.2024.07.001] [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: 03/11/2024] [Revised: 05/23/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024]
Abstract
Peptostreptococcus stomatis (P. stomatis) is enriched in colorectal cancer (CRC), but its causality and translational implications in CRC are unknown. Here, we show that P. stomatis accelerates colonic tumorigenesis in ApcMin/+ and azoxymethane/dextran sodium sulfate (AOM-DSS) models by inducing cell proliferation, suppressing apoptosis, and impairing gut barrier function. P. stomatis adheres to CRC cells through its surface protein fructose-1,6-bisphosphate aldolase (FBA) that binds to the integrin α6/β4 receptor on CRC cells, leading to the activation of ERBB2 and the downstream MEK-ERK-p90 cascade. Blockade of the FBA-integrin α6/β4 abolishes ERBB2-mitogen-activated protein kinase (MAPK) activation and the protumorigenic effect of P. stomatis. P. stomatis-driven ERBB2 activation bypasses receptor tyrosine kinase (RTK) blockade by EGFR inhibitors (cetuximab, erlotinib), leading to drug resistance in xenograft and spontaneous CRC models of KRAS-wild-type CRC. P. stomatis also abrogates BRAF inhibitor (vemurafenib) efficacy in BRAFV600E-mutant CRC xenografts. Thus, we identify P. stomatis as an oncogenic bacterium and a contributory factor for non-responsiveness to RTK inhibitors in CRC.
Collapse
Affiliation(s)
- Pingmei Huang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Fenfen Ji
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Alvin Ho-Kwan Cheung
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kaili Fu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qiming Zhou
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiao Ding
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Danyu Chen
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yufeng Lin
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Luyao Wang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ying Jiao
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Eagle S H Chu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Chi Chun Wong
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
41
|
Khandan V, Boerkamp VJP, Chiechi RC, Hohlbein J, Mathwig K. Addressing spatiotemporal signal variations in pair correlation function analysis. Biophys J 2024:S0006-3495(24)00524-1. [PMID: 39113360 DOI: 10.1016/j.bpj.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/22/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024] Open
Abstract
Fluorescence correlation spectroscopy (FCS) is a cornerstone technique in optical microscopy to measure, for example, the concentration and diffusivity of fluorescent emitters and biomolecules in solution. The application of FCS to complex biological systems, however, is fraught with inherent intricacies that impair the interpretation of correlation patterns. Critical among these intricacies are temporal variations beyond diffusion in the quantity, intensity, and spatial distribution of fluorescent emitters. These variations introduce distortions into correlated intensity data, thus compromising the accuracy and reproducibility of the analysis. This issue is accentuated in imaging-based approaches such as pair correlation function (pCF) analysis due to their broader regions of interest compared with point-detector-based approaches. Despite ongoing developments in FCS, attention to systems characterized by a spatiotemporal-dependent probability distribution function (ST-PDF) has been lacking. To address this knowledge gap, we developed a new analytical framework for ST-PDF systems that introduces a dual-timescale model function within the conventional pCF analysis. Our approach selectively differentiates the signals associated with rapid processes, such as particle diffusion, from signals stemming from spatiotemporal variations in the distribution of fluorescent emitters occurring at extended delay timescales. To corroborate our approach, we conducted proof-of-concept experiments on an ST-PDF system, wherein the, initially, uniform distribution of fluorescent microspheres within a microfluidic channel changes into a localized accumulation of microspheres over time. Our framework is offering a comprehensive solution for investigating various phenomena such as biomolecular binding, sedimentation, and particle accumulation.
Collapse
Affiliation(s)
- Vahid Khandan
- University of Groningen, Groningen Research Institute of Pharmacy, Pharmaceutical Analysis, Groningen, the Netherlands
| | - Vincent J P Boerkamp
- Laboratory of Biophysics, Wageningen University & Research, Wageningen, the Netherlands
| | - Ryan C Chiechi
- Department of Chemistry & Organic and Carbon Electronics Laboratory, North Carolina State University, Raleigh, North Carolina
| | - Johannes Hohlbein
- Laboratory of Biophysics, Wageningen University & Research, Wageningen, the Netherlands; Microspectroscopy Research Facility, Wageningen University & Research, Wageningen, the Netherlands.
| | - Klaus Mathwig
- University of Groningen, Groningen Research Institute of Pharmacy, Pharmaceutical Analysis, Groningen, the Netherlands; imec within OnePlanet Research Center, Wageningen, the Netherlands.
| |
Collapse
|
42
|
Mata Calidonio J, Maddox AI, Hamad-Schifferli K. A novel immunoassay technique using principal component analysis for enhanced detection of emerging viral variants. LAB ON A CHIP 2024; 24:3985-3995. [PMID: 39046406 DOI: 10.1039/d4lc00505h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
Rapid diagnostics are critical infectious disease tools that are designed to detect a known biomarker using antibodies specific to that biomarker. However, a way to detect unknown disease variants has not yet been achieved in a paper test format. We describe here a route to make an adaptable paper immunoassay that can detect an unknown biomarker, demonstrating it on SARS-CoV-2 variants. The immunoassay repurposes cross reactive antibodies raised against the alpha variant. Gold nanoparticles of two different colors conjugated to two different antibodies create a colorimetric signal, and machine learning of the resulting colorimetric pattern is used to train the assay to discriminate between variants of alpha and Omicron BA.5. By using principal component analysis, the colorimetric test patterns can pick up and discriminate an unknown variant that it has not encountered before, Omicron BA.1. The test has an accuracy of 100% and a potential calculated discriminatory power of 900. We show that it can be used adaptively and that it can be used to pick up emerging variants without the need to raise new antibodies.
Collapse
Affiliation(s)
| | - Arianna I Maddox
- Department of Biology, University of Massachusetts Boston, Boston, MA, USA
| | - Kimberly Hamad-Schifferli
- Department of Engineering, University of Massachusetts Boston, Boston, MA, USA.
- School for the Environment, University of Massachusetts Boston, Boston, MA, USA
| |
Collapse
|
43
|
Jagilinki BP, Willis MA, Mus F, Sharma R, Pellows LM, Mulder DW, Yang ZY, Seefeldt LC, King PW, Dukovic G, Peters JW. Microscale Thermophoresis (MST) as a Tool to Study Binding Interactions of Oxygen-Sensitive Biohybrids. Bio Protoc 2024; 14:e5041. [PMID: 39131194 PMCID: PMC11309957 DOI: 10.21769/bioprotoc.5041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 08/13/2024] Open
Abstract
Microscale thermophoresis (MST) is a technique used to measure the strength of molecular interactions. MST is a thermophoretic-based technique that monitors the change in fluorescence associated with the movement of fluorescent-labeled molecules in response to a temperature gradient triggered by an IR LASER. MST has advantages over other approaches for examining molecular interactions, such as isothermal titration calorimetry, nuclear magnetic resonance, biolayer interferometry, and surface plasmon resonance, requiring a small sample size that does not need to be immobilized and a high-sensitivity fluorescence detection. In addition, since the approach involves the loading of samples into capillaries that can be easily sealed, it can be adapted to analyze oxygen-sensitive samples. In this Bio-protocol, we describe the troubleshooting and optimization we have done to enable the use of MST to examine protein-protein interactions, protein-ligand interactions, and protein-nanocrystal interactions. The salient elements in the developed procedures include 1) loading and sealing capabilities in an anaerobic chamber for analysis using a NanoTemper MST located on the benchtop in air, 2) identification of the optimal reducing agents compatible with data acquisition with effective protection against trace oxygen, and 3) the optimization of data acquisition and analysis procedures. The procedures lay the groundwork to define the determinants of molecular interactions in these technically demanding systems. Key features • Established procedures for loading and sealing tubes in an anaerobic chamber for subsequent analysis. • Sodium dithionite (NaDT) could easily be substituted with one electron-reduced 1,1'-bis(3-sulfonatopropyl)-4,4'-bipyridinium [(SPr)2V•] to perform sensitive biophysical assays on oxygen-sensitive proteins like the MoFe protein. • Established MST as an experimental tool to quantify binding affinities in novel enzyme-quantum dot biohybrid complexes that are extremely oxygen-sensitive.
Collapse
Affiliation(s)
- Bhanu P. Jagilinki
- Department of Chemistry and Biochemistry, The University of Oklahoma, Norman, OK, USA
| | - Mark A. Willis
- Institute of Biological Chemistry, Washington State University, Pullman, WA, USA
| | - Florence Mus
- Department of Chemistry and Biochemistry, The University of Oklahoma, Norman, OK, USA
| | - Ritika Sharma
- Department of Chemistry and Biochemistry, The University of Oklahoma, Norman, OK, USA
| | - Lauren M. Pellows
- Department of Chemistry, University of Colorado Boulder, Boulder, CO, USA
| | - David W. Mulder
- Biosciences Center, National Renewable Energy Laboratory, Golden, CO, USA
| | - Zhi-Yong Yang
- Department of Chemistry and Biochemistry, Utah State University, Logan, UT, USA
| | - Lance C. Seefeldt
- Department of Chemistry and Biochemistry, Utah State University, Logan, UT, USA
| | - Paul W. King
- Biosciences Center, National Renewable Energy Laboratory, Golden, CO, USA
| | - Gordana Dukovic
- Department of Chemistry, University of Colorado Boulder, Boulder, CO, USA
- Materials Science and Engineering and Renewable and Sustainable Energy Institute (RASEI), University of Colorado Boulder, Boulder, CO, USA
| | - John W. Peters
- Department of Chemistry and Biochemistry, The University of Oklahoma, Norman, OK, USA
| |
Collapse
|
44
|
Zhang T, Liu Y, Cao J, Liu Y, Hao L, Lin K, Yi H. Exploration of Novel Plasmin Inhibitor from β-Lactoglobulin for Enhancing the Storage Stability of UHT Milk. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:17041-17050. [PMID: 39024493 DOI: 10.1021/acs.jafc.4c04986] [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: 07/20/2024]
Abstract
Plasmin-induced protein hydrolysis significantly compromises the stability of ultrahigh-temperature (UHT) milk. β-Lactoglobulin (β-Lg) was observed to inhibit plasmin activity, suggesting that there were active sites as plasmin inhibitors in β-Lg. Herein, plasmin inhibitory peptides were explored from β-Lg using experimental and computational techniques. The results revealed that increased denaturation of β-Lg enhanced its affinity for plasmin, leading to a stronger inhibition of plasmin activity. Molecular dynamics simulations indicated that electrostatic and van der Waals forces were the primary binding forces in the β-Lg/plasmin complex. Denatured β-Lg increased hydrogen bonding and reduced the binding energy with plasmin. The sites of plasmin bound to β-Lg were His624, Asp667, and Ser762. Four plasmin inhibitory peptides, QTMKGLDI, EKTKIPAV, TDYKKYLL, and CLVRTPEV, were identified from β-Lg based on binding sites. These peptides effectively inhibited plasmin activity and enhanced the UHT milk stability. This study provided new insights into the development of novel plasmin inhibitors to improve the stability of UHT milk.
Collapse
Affiliation(s)
- Tai Zhang
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong Province 266003, China
- Food Laboratory of Zhongyuan, Luohe, Henan Province 462300, China
| | - Yisuo Liu
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong Province 266003, China
- Food Laboratory of Zhongyuan, Luohe, Henan Province 462300, China
| | - Jiayuan Cao
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong Province 266003, China
| | - Yinxue Liu
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong Province 266003, China
| | - Linlin Hao
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong Province 266003, China
| | - Kai Lin
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong Province 266003, China
| | - Huaxi Yi
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong Province 266003, China
- Food Laboratory of Zhongyuan, Luohe, Henan Province 462300, China
| |
Collapse
|
45
|
Peixoto ML, Madan E. Unraveling the complexity: Advanced methods in analyzing DNA, RNA, and protein interactions. Adv Cancer Res 2024; 163:251-302. [PMID: 39271265 DOI: 10.1016/bs.acr.2024.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Exploring the intricate interplay within and between nucleic acids, as well as their interactions with proteins, holds pivotal significance in unraveling the molecular complexities steering cancer initiation and progression. To investigate these interactions, a diverse array of highly specific and sensitive molecular techniques has been developed. The selection of a particular technique depends on the specific nature of the interactions. Typically, researchers employ an amalgamation of these different techniques to obtain a comprehensive and holistic understanding of inter- and intramolecular interactions involving DNA-DNA, RNA-RNA, DNA-RNA, or protein-DNA/RNA. Examining nucleic acid conformation reveals alternative secondary structures beyond conventional ones that have implications for cancer pathways. Mutational hotspots in cancer often lie within sequences prone to adopting these alternative structures, highlighting the importance of investigating intra-genomic and intra-transcriptomic interactions, especially in the context of mutations, to deepen our understanding of oncology. Beyond these intramolecular interactions, the interplay between DNA and RNA leads to formations like DNA:RNA hybrids (known as R-loops) or even DNA:DNA:RNA triplex structures, both influencing biological processes that ultimately impact cancer. Protein-nucleic acid interactions are intrinsic cellular phenomena crucial in both normal and pathological conditions. In particular, genetic mutations or single amino acid variations can alter a protein's structure, function, and binding affinity, thus influencing cancer progression. It is thus, imperative to understand the differences between wild-type (WT) and mutated (MT) genes, transcripts, and proteins. The review aims to summarize the frequently employed methods and techniques for investigating interactions involving nucleic acids and proteins, highlighting recent advancements and diverse adaptations of each technique.
Collapse
Affiliation(s)
- Maria Leonor Peixoto
- Champalimaud Center for the Unknown, Lisbon, Portugal; Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Esha Madan
- Department of Surgery, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, United States; VCU Institute of Molecular Medicine, Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
46
|
Brahma R, Raghuraman H. Characterization of a novel MgtE homolog and its structural dynamics in membrane mimetics. Biophys J 2024; 123:1968-1983. [PMID: 38042987 PMCID: PMC11309985 DOI: 10.1016/j.bpj.2023.11.3402] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/03/2023] [Accepted: 11/30/2023] [Indexed: 12/04/2023] Open
Abstract
Magnesium (Mg2+) is the most abundant divalent cation in the cell and is critical for numerous cellular processes. Despite its importance, the mechanisms of intracellular Mg2+ transport and its regulation are poorly understood. MgtE is the main Mg2+ transport system in almost half of bacterial species and is an ortholog of mammalian SLC41A1 transporters, which are implicated in neurodegenerative diseases and cancer. To date, only MgtE from Thermus thermophilus (MgtETT) has been extensively characterized, mostly in detergent micelles, and gating-related structural dynamics in biologically relevant membranes are scarce. The MgtE homolog from Bacillus firmus (MgtEBF) is unique since it lacks the entire Mg2+-sensing N-domain but has conserved structural motifs in the TM-domain for Mg2+ transport. In this work, we have successfully purified this novel homolog in a stable and functional form, and ColabFold structure prediction analysis suggests a homodimer. Further, microscale thermophoresis experiments show that MgtEBF binds Mg2+ and ATP, similar to MgtETT. Importantly, we show that, despite lacking the N-domain, MgtEBF mediates Mg2+ transport function in the presence of an inwardly directed Mg2+ gradient in reconstituted proteoliposomes. Furthermore, comparison of the organization and dynamics of Trp residues in the TM-domain of MgtEBF in membrane mimetics, in apo- and Mg2+-bound forms, suggests that the cytoplasmic binding of Mg2+ might involve modest gating-related conformational changes at the TM-domain. Overall, our results show that the gating-related structural dynamics (hydration dynamics, conformational heterogeneity) of the full-length MgtEBF is significantly changed in functionally pertinent membrane environment, emphasizing the importance of lipid-protein interactions in MgtE gating mechanisms.
Collapse
Affiliation(s)
- Rupasree Brahma
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, Training School Complex, Mumbai, India
| | - H Raghuraman
- Crystallography and Molecular Biology Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, Training School Complex, Mumbai, India.
| |
Collapse
|
47
|
Zhang J, Liu B, Chen H, Zhang L, Jiang X. Application and Method of Surface Plasmon Resonance Technology in the Preparation and Characterization of Biomedical Nanoparticle Materials. Int J Nanomedicine 2024; 19:7049-7069. [PMID: 39011388 PMCID: PMC11249113 DOI: 10.2147/ijn.s468695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/02/2024] [Indexed: 07/17/2024] Open
Abstract
Surface Plasmon Resonance (SPR) technology, as a powerful analytical tool, plays a crucial role in the preparation, performance evaluation, and biomedical applications of nanoparticles due to its real-time, label-free, and highly sensitive detection capabilities. In the nanoparticle preparation process, SPR technology can monitor synthesis reactions and surface modifications in real-time, optimizing preparation techniques and conditions. SPR enables precise measurement of interactions between nanoparticles and biomolecules, including binding affinities and kinetic parameters, thereby assessing nanoparticle performance. In biomedical applications, SPR technology is extensively used in the study of drug delivery systems, biomarker detection for disease diagnosis, and nanoparticle-biomolecule interactions. This paper reviews the latest advancements in SPR technology for nanoparticle preparation, performance evaluation, and biomedical applications, discussing its advantages and challenges in biomedical applications, and forecasting future development directions.
Collapse
Affiliation(s)
- Jingyao Zhang
- Core Facilities of West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Beibei Liu
- Core Facilities of West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Hongying Chen
- Core Facilities of West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Lingshu Zhang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xia Jiang
- Division of Biliary Tract Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People's Republic of China
| |
Collapse
|
48
|
Lühmann KL, Seemann S, Martinek N, Ostendorp S, Kehr J. The glycine-rich domain of GRP7 plays a crucial role in binding long RNAs and facilitating phase separation. Sci Rep 2024; 14:16018. [PMID: 38992080 PMCID: PMC11239674 DOI: 10.1038/s41598-024-66955-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 07/05/2024] [Indexed: 07/13/2024] Open
Abstract
Microscale thermophoresis (MST) is a well-established method to quantify protein-RNA interactions. In this study, we employed MST to analyze the RNA binding properties of glycine-rich RNA binding protein 7 (GRP7), which is known to have multiple biological functions related to its ability to bind different types of RNA. However, the exact mechanism of GRP7's RNA binding is not fully understood. While the RNA-recognition motif of GRP7 is known to be involved in RNA binding, the glycine-rich region (known as arginine-glycine-glycine-domain or RGG-domain) also influences this interaction. To investigate to which extend the RGG-domain of GRP7 is involved in RNA binding, mutation studies on putative RNA interacting or modulating sites were performed. In addition to MST experiments, we examined liquid-liquid phase separation of GRP7 and its mutants, both with and without RNA. Furthermore, we systemically investigated factors that might affect RNA binding selectivity of GRP7 by testing RNAs of different sizes, structures, and modifications. Consequently, our study revealed that GRP7 exhibits a high affinity for a variety of RNAs, indicating a lack of pronounced selectivity. Moreover, we established that the RGG-domain plays a crucial role in binding longer RNAs and promoting phase separation.
Collapse
Affiliation(s)
- Kim Lara Lühmann
- Department of Biology, Molecular Plant Genetics, Institute of Plant Science and Microbiology, Universität Hamburg, Hamburg, Germany
| | - Silja Seemann
- Department of Biology, Molecular Plant Genetics, Institute of Plant Science and Microbiology, Universität Hamburg, Hamburg, Germany
| | - Nina Martinek
- Department of Biology, Molecular Plant Genetics, Institute of Plant Science and Microbiology, Universität Hamburg, Hamburg, Germany
| | - Steffen Ostendorp
- Department of Biology, Molecular Plant Genetics, Institute of Plant Science and Microbiology, Universität Hamburg, Hamburg, Germany
| | - Julia Kehr
- Department of Biology, Molecular Plant Genetics, Institute of Plant Science and Microbiology, Universität Hamburg, Hamburg, Germany.
| |
Collapse
|
49
|
Atxabal U, Fernández A, Moure MJ, Sobczak K, Nycholat C, Almeida-Marrero V, Oyenarte I, Paulson JC, de la Escosura A, Torres T, Reichardt NC, Jiménez-Barbero J, Ereño-Orbea J. Quantifying Siglec-sialylated ligand interactions: a versatile 19F-T 2 CPMG filtered competitive NMR displacement assay. Chem Sci 2024; 15:10612-10624. [PMID: 38994400 PMCID: PMC11234860 DOI: 10.1039/d4sc01723d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/08/2024] [Indexed: 07/13/2024] Open
Abstract
Sialic-acid-binding immunoglobulin-like lectins (Siglecs) are integral cell surface proteins crucial for the regulation of immune responses and the maintenance of immune tolerance through interactions with sialic acids. Siglecs recognize sialic acid moieties, usually found at the end of N-glycan and O-glycan chains. However, the different Siglecs prefer diverse presentations of the recognized sialic acid, depending on the type of glycosidic linkage used to link to the contiguous Gal/GalNAc or sialic acid moieties. This fact, together with possible O- or N-substitutions at the recognized glycan epitope significantly influences their roles in various immune-related processes. Understanding the molecular details of Siglec-sialoglycan interactions is essential for unraveling their specificities and for the development of new molecules targeting these receptors. While traditional biophysical methods like isothermal titration calorimetry (ITC) have been utilized to measure binding between lectins and glycans, contemporary techniques such as surface plasmon resonance (SPR), microscale thermophoresis (MST), and biolayer interferometry (BLI) offer improved throughput. However, these methodologies require chemical modification and immobilization of at least one binding partner, which can interfere the recognition between the lectin and the ligand. Since Siglecs display a large range of dissociation constants, depending on the (bio)chemical nature of the interacting partner, a general and robust method that could monitor and quantify binding would be highly welcomed. Herein, we propose the application of an NMR-based a competitive displacement assay, grounded on 19F T2-relaxation NMR and on the design, synthesis, and use of a strategic spy molecule, to assess and quantify sialoside ligand binding to Siglecs. We show that the use of this specific approach allows the quantification of Siglec binding for natural and modified sialosides, multivalent sialosides, and sialylated glycoproteins in solution, which differ in binding affinities in more than two orders of magnitude, thus providing invaluable insights into sialoglycan-mediated interactions.
Collapse
Affiliation(s)
- Unai Atxabal
- Chemical Glycobiology Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA) 48160 Derio Bizkaia Spain
| | - Andrea Fernández
- Chemical Glycobiology Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA) 48160 Derio Bizkaia Spain
- Glycotechnology Laboratory, CIC biomaGUNE Paseo Miramon 194 San Sebastian 20014 Spain
| | - Maria Jesús Moure
- Chemical Glycobiology Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA) 48160 Derio Bizkaia Spain
| | - Klaudia Sobczak
- Chemical Glycobiology Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA) 48160 Derio Bizkaia Spain
| | - Corwin Nycholat
- Departments of Molecular Medicine and Immunology & Microbiology, The Scripps Research Institute 10550 North Torrey Pines Road La Jolla California 92037 USA
| | - Verónica Almeida-Marrero
- Department of Organic Chemistry, Universidad Autónoma de Madrid C/Francisco Tomás y Valiente 7 28049 Madrid Spain
- Institute for Advanced Research in Chemical Sciences (IAdChem), Universidad Autónoma de Madrid C/Francisco Tomás y Valiente 7 28049 Madrid Spain
| | - Iker Oyenarte
- Chemical Glycobiology Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA) 48160 Derio Bizkaia Spain
| | - James C Paulson
- Departments of Molecular Medicine and Immunology & Microbiology, The Scripps Research Institute 10550 North Torrey Pines Road La Jolla California 92037 USA
| | - Andrés de la Escosura
- Department of Organic Chemistry, Universidad Autónoma de Madrid C/Francisco Tomás y Valiente 7 28049 Madrid Spain
- Institute for Advanced Research in Chemical Sciences (IAdChem), Universidad Autónoma de Madrid C/Francisco Tomás y Valiente 7 28049 Madrid Spain
| | - Tomás Torres
- Department of Organic Chemistry, Universidad Autónoma de Madrid C/Francisco Tomás y Valiente 7 28049 Madrid Spain
- Institute for Advanced Research in Chemical Sciences (IAdChem), Universidad Autónoma de Madrid C/Francisco Tomás y Valiente 7 28049 Madrid Spain
- Instituto Madrileño de Estudios Avanzados (IMDEA)-Nanociencia C/Faraday 9 28049 Madrid Spain
| | - Niels C Reichardt
- Glycotechnology Laboratory, CIC biomaGUNE Paseo Miramon 194 San Sebastian 20014 Spain
- CIBER-BBN Paseo Miramon 194 San Sebastian 20014 Spain
| | - Jesús Jiménez-Barbero
- Chemical Glycobiology Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA) 48160 Derio Bizkaia Spain
- Ikerbasque, Basque Foundation for Science Bilbao Spain
- Department of Organic & Inorganic Chemistry, Faculty of Science and Technology, University of the Basque Country, EHU-UPV 48940 Leioa Bizkaia Spain
- Centro de Investigacion Biomedica en Red de Enfermedades Respiratorias 28029 Madrid Spain
| | - June Ereño-Orbea
- Chemical Glycobiology Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA) 48160 Derio Bizkaia Spain
- Ikerbasque, Basque Foundation for Science Bilbao Spain
| |
Collapse
|
50
|
Šoltysová M, Škerlová J, Pachl P, Škubník K, Fábry M, Sieglová I, Farolfi M, Grishkovskaya I, Babiak M, Nováček J, Krásný L, Řezáčová P. Structural characterization of two prototypical repressors of SorC family reveals tetrameric assemblies on DNA and mechanism of function. Nucleic Acids Res 2024; 52:7305-7320. [PMID: 38842936 PMCID: PMC11229326 DOI: 10.1093/nar/gkae434] [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: 12/05/2023] [Revised: 04/16/2024] [Accepted: 05/22/2024] [Indexed: 07/09/2024] Open
Abstract
The SorC family of transcriptional regulators plays a crucial role in controlling the carbohydrate metabolism and quorum sensing. We employed an integrative approach combining X-ray crystallography and cryo-electron microscopy to investigate architecture and functional mechanism of two prototypical representatives of two sub-classes of the SorC family: DeoR and CggR from Bacillus subtilis. Despite possessing distinct DNA-binding domains, both proteins form similar tetrameric assemblies when bound to their respective DNA operators. Structural analysis elucidates the process by which the CggR-regulated gapA operon is derepressed through the action of two effectors: fructose-1,6-bisphosphate and newly confirmed dihydroxyacetone phosphate. Our findings provide the first comprehensive understanding of the DNA binding mechanism of the SorC-family proteins, shedding new light on their functional characteristics.
Collapse
Affiliation(s)
- Markéta Šoltysová
- Structural Biology, Institute of Organic Chemistry and Biochemistry of Czech Academy of Sciences, Prague, 166 10, Czechia
| | - Jana Škerlová
- Structural Biology, Institute of Organic Chemistry and Biochemistry of Czech Academy of Sciences, Prague, 166 10, Czechia
| | - Petr Pachl
- Structural Biology, Institute of Organic Chemistry and Biochemistry of Czech Academy of Sciences, Prague, 166 10, Czechia
| | - Karel Škubník
- CryoElectron Microscopy and Tomography Core Facility, Central European Institute of Technology, Brno, 601 77, Czechia
| | - Milan Fábry
- Structural Biology, Institute of Organic Chemistry and Biochemistry of Czech Academy of Sciences, Prague, 166 10, Czechia
| | - Irena Sieglová
- Structural Biology, Institute of Organic Chemistry and Biochemistry of Czech Academy of Sciences, Prague, 166 10, Czechia
| | - Martina Farolfi
- Laboratory of Microbial Genetics and Gene Expression, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 142 20, Czechia
| | - Irina Grishkovskaya
- Research Institute of Molecular Pathology, Campus-ViennaBiocenter 1, 1030 Vienna, Austria
| | - Michal Babiak
- CryoElectron Microscopy and Tomography Core Facility, Central European Institute of Technology, Brno, 601 77, Czechia
| | - Jiří Nováček
- CryoElectron Microscopy and Tomography Core Facility, Central European Institute of Technology, Brno, 601 77, Czechia
| | - Libor Krásný
- Laboratory of Microbial Genetics and Gene Expression, Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, Prague 142 20, Czechia
| | - Pavlína Řezáčová
- Structural Biology, Institute of Organic Chemistry and Biochemistry of Czech Academy of Sciences, Prague, 166 10, Czechia
| |
Collapse
|