1
|
Aranda-García D, Stepniewski TM, Torrens-Fontanals M, García-Recio A, Lopez-Balastegui M, Medel-Lacruz B, Morales-Pastor A, Peralta-García A, Dieguez-Eceolaza M, Sotillo-Nuñez D, Ding T, Drabek M, Jacquemard C, Jakowiecki J, Jespers W, Jiménez-Rosés M, Jun-Yu-Lim V, Nicoli A, Orzel U, Shahraki A, Tiemann JKS, Ledesma-Martin V, Nerín-Fonz F, Suárez-Dou S, Canal O, Pándy-Szekeres G, Mao J, Gloriam DE, Kellenberger E, Latek D, Guixà-González R, Gutiérrez-de-Terán H, Tikhonova IG, Hildebrand PW, Filizola M, Babu MM, Di Pizio A, Filipek S, Kolb P, Cordomi A, Giorgino T, Marti-Solano M, Selent J. Large scale investigation of GPCR molecular dynamics data uncovers allosteric sites and lateral gateways. Nat Commun 2025; 16:2020. [PMID: 40016203 PMCID: PMC11868581 DOI: 10.1038/s41467-025-57034-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: 05/15/2024] [Accepted: 02/07/2025] [Indexed: 03/01/2025] Open
Abstract
G protein-coupled receptors (GPCRs) constitute a functionally diverse protein family and are targets for a broad spectrum of pharmaceuticals. Technological progress in X-ray crystallography and cryogenic electron microscopy has enabled extensive, high-resolution structural characterisation of GPCRs in different conformational states. However, as highly dynamic events underlie GPCR signalling, a complete understanding of GPCR functionality requires insights into their conformational dynamics. Here, we present a large dataset of molecular dynamics simulations covering 60% of currently available GPCR structures. Our analysis reveals extensive local "breathing" motions of the receptor on a nano- to microsecond timescale and provides access to numerous previously unexplored receptor conformational states. Furthermore, we reveal that receptor flexibility impacts the shape of allosteric drug binding sites, which frequently adopt partially or completely closed states in the absence of a molecular modulator. We demonstrate that exploring membrane lipid dynamics and their interaction with GPCRs is an efficient approach to expose such hidden allosteric sites and even lateral ligand entrance gateways. The obtained insights and generated dataset on conformations, allosteric sites and lateral entrance gates in GPCRs allows us to better understand the functionality of these receptors and opens new therapeutic avenues for drug-targeting strategies.
Collapse
Affiliation(s)
- David Aranda-García
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Tomasz Maciej Stepniewski
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
- InterAx Biotech AG, Villigen, Switzerland
| | - Mariona Torrens-Fontanals
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
- Acellera Labs, Barcelona, Spain
| | - Adrian García-Recio
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Marta Lopez-Balastegui
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Brian Medel-Lacruz
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Adrián Morales-Pastor
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | | | - Miguel Dieguez-Eceolaza
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - David Sotillo-Nuñez
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
| | - Tianyi Ding
- School of Pharmacy, Medical Biology Centre, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Matthäus Drabek
- Department of Pharmaceutical Chemistry, University of Marburg, Marburg, Germany
| | - Célien Jacquemard
- Laboratoire d'Innovation Thérapeutique, University of Strasbourg, Strasbourg, France
| | - Jakub Jakowiecki
- Faculty of Chemistry, University of Warsaw, Warsaw, Poland
- Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Willem Jespers
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Mireia Jiménez-Rosés
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, Birmingham and Nottingham, Midlands, UK
- Sygnature Discovery Ltd., Nottingham, UK
| | - Víctor Jun-Yu-Lim
- Department of Pharmaceutical Chemistry, University of Marburg, Marburg, Germany
| | - Alessandro Nicoli
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
| | - Urszula Orzel
- Faculty of Chemistry, University of Warsaw, Warsaw, Poland
- Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Aida Shahraki
- Department of Pharmaceutical Chemistry, University of Marburg, Marburg, Germany
| | - Johanna K S Tiemann
- Medizinische Fakultät, Institut für Medizinische Physik und Biophysik, Universität Leipzig, Leipzig, Germany
| | - Vicente Ledesma-Martin
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
| | - Francho Nerín-Fonz
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
| | - Sergio Suárez-Dou
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
| | - Oriol Canal
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain
| | - Gáspár Pándy-Szekeres
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Medicinal Chemistry Research Group, HUN-REN Research Center for Natural Sciences, Budapest, Hungary
| | - Jiafei Mao
- Beijing National Laboratory for Molecular Sciences (BNLMS) and Center for Physicochemical Analysis and Measurement, Institute of Chemistry Chinese Academy of Science (ICCAS), Beijing, China
| | - David E Gloriam
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Esther Kellenberger
- Laboratoire d'Innovation Thérapeutique, University of Strasbourg, Strasbourg, France
| | - Dorota Latek
- Faculty of Chemistry, University of Warsaw, Warsaw, Poland
| | - Ramon Guixà-González
- Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Hugo Gutiérrez-de-Terán
- Department of Cell and Molecular Biology, Uppsala University, Biomedical Center, Uppsala, Sweden
- Research Center in Nanomaterials and Nanotechnology (CINN/CSIC) and Health Institute of Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Irina G Tikhonova
- School of Pharmacy, Medical Biology Centre, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Peter W Hildebrand
- Institute of Medical Physics and Biophysics, Medical University Leipzig, Leipzig, Sachsen, Germany
| | - Marta Filizola
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - M Madan Babu
- Department of Structural Biology and Center of Excellence for Data Driven Discovery, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Antonella Di Pizio
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- Chemoinformatics and Protein Modelling, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | | | - Peter Kolb
- Department of Pharmaceutical Chemistry, University of Marburg, Marburg, Germany
| | - Arnau Cordomi
- Department of Biochemistry and Molecular Biology, Faculty of Biosciences, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
| | - Toni Giorgino
- Institute of Biophysics (IBF-CNR), National Research Council of Italy, Milano, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | | | - Jana Selent
- Department of Medicine and Life Sciences, Pompeu Fabra University (UPF), Barcelona, Spain.
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain.
| |
Collapse
|
2
|
Huang L, Huhulea E, Aifuwa E, Frishman WH, Aronow WS. Sugar-Free but Not Risk-Free? Exploring Artificial Sweeteners and Cardiovascular Disease. Cardiol Rev 2025:00045415-990000000-00425. [PMID: 39969176 DOI: 10.1097/crd.0000000000000873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
The consumption of artificial sweeteners has significantly increased globally, particularly as a substitute for sugar for the management of conditions such as diabetes and obesity, which are significant risk factors for cardiovascular disease. Despite their widespread use, the health impacts of artificial sweeteners remain contentious. Research has suggested that certain sweeteners may contribute to systemic inflammation, endothelial dysfunction, and disruptions in gut microbiota, potentially altering glucose metabolism and exacerbating metabolic conditions such as diabetes and obesity. However, other studies highlight potential benefits, such as weight control and improved glucose tolerance. Still, the long-term safety of artificial sweeteners, particularly with chronic consumption, remains uncertain. This literature review explores the cardiovascular risks associated with various artificial sweeteners, focusing on the 6 US Food and Drug Administration-approved nonnutritive sweeteners, aspartame, sucralose, saccharin, acesulfame K, cyclamate, and neotame, as well as nutritive sweeteners such as polyols (erythritol, xylitol, sorbitol, and maltitol). Ongoing research, including in vitro, animal, and clinical studies, aims to clarify the long-term cardiovascular and metabolic implications of artificial sweeteners and assess the safety of their widespread use across diverse populations.
Collapse
Affiliation(s)
- Lillian Huang
- Department of Medicine, New York Medical College, Valhalla, NY
| | - Ellen Huhulea
- Department of Medicine, New York Medical College, Valhalla, NY
| | - Eseiwi Aifuwa
- Department of Medicine, New York Medical College, Valhalla, NY
| | | | - Wilbert S Aronow
- Department of Medicine, New York Medical College, Valhalla, NY
- Departments of Cardiology and Medicine, Westchester Medical Center, Valhalla, NY
| |
Collapse
|
3
|
Wei F, Luo L, Wang X, Luo W, Wu F, Tian S, Qin Y, Zeng L. The masking mechanism of catechin to the sweet taste of phloridzin. Food Chem 2025; 464:141756. [PMID: 39461313 DOI: 10.1016/j.foodchem.2024.141756] [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: 05/03/2024] [Revised: 09/29/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024]
Abstract
Our previous study speculated that decreased levels of catechin were associated with heightened sweetness in Camellia nanchuanica black tea (NCBT). In this study, we found that catechin has the capacity to obscure the sweet taste of phloridzin and verified the above speculation. To delve deeper into this masking effect, we examined various concentrations of catechin (cat-10cat) and phloridzin (phl-10phl), and revealed catechin (cat-10cat) significantly reduced the sweetness of 10phl. Therefore, catechin (cat-10cat) and 10phl combinations were selected to investigate the impact on sweet taste receptor cells. The results showed increasing concentrations of catechin inhibited the calcium signal of the phloridzin-catechin solution, attributing to lower stability of phloridzin and Taste 1 Receptor Member 3 (one subunit of sweet taste receptor proteins) after addition catechin. Finally, 10phl/10cat significantly activated the central and prefrontal regions generating more bitter tastes and negative emotions which also probably contributed to mask sweet tastes of phloridzin.
Collapse
Affiliation(s)
- Fang Wei
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City/College of Food Science, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China; Chongqing Tea Technology and Innovation Center, Chongqing, China
| | - Liyong Luo
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City/College of Food Science, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China; Chongqing Tea Technology and Innovation Center, Chongqing, China
| | - Xi Wang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City/College of Food Science, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China; Chongqing Tea Technology and Innovation Center, Chongqing, China
| | - Wei Luo
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City/College of Food Science, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China; Chongqing Tea Technology and Innovation Center, Chongqing, China
| | - Fan Wu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City/College of Food Science, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China; Chongqing Tea Technology and Innovation Center, Chongqing, China
| | - Shiyi Tian
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Yumei Qin
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Liang Zeng
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City/College of Food Science, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China; Chongqing Tea Technology and Innovation Center, Chongqing, China.
| |
Collapse
|
4
|
Yu H, Ao T, Mao H, Liu J, Chen C, Tian H. Sweet-enhancing effect of coolant agent menthol evaluated via sensory analysis and molecular modeling. Food Chem X 2025; 26:102337. [PMID: 40123878 PMCID: PMC11930197 DOI: 10.1016/j.fochx.2025.102337] [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: 01/10/2025] [Revised: 02/18/2025] [Accepted: 02/26/2025] [Indexed: 03/25/2025] Open
Abstract
Responding to global trends favoring low-sugar diets, this study explored the potential of menthol, a cooling agent, to enhance sweet taste perception through integrated sensory evaluations and molecular modeling. The results of static sensory evaluation (recognition threshold determination, paired comparison test and 15 cm-linear scale) and dynamic sensory analysis indicated that menthol lowered sweetness threshold of HFCS (from 5.98 g/L to 5.02 g/L), while intensifying maximum sweetness intensity and prolonging the duration of sweetness. Sensory analysis identified optimal sweet enhancement at 0.004-0.030 g/L menthol concentrations, while 0.060 g/L caused sweetness suppression through intensified cooling/bitter sensations. Molecular modeling comparing T1R2/T1R3-Glu/Fru system and T1R2/T1R3-Glu/Fru/Men system elucidated that the addition of menthol increased the number of hotspot residues in protein-sugars binding and stabilized interactions by occupying sites near sugar active sites, maintaining the Venus Flytrap Domain in its closed, activated configuration. These findings demonstrated the underlying contribution menthol made to sweet enhancement and sugar reduction.
Collapse
Affiliation(s)
- Haiyan Yu
- Department of Food Science and Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Ting Ao
- Department of Food Science and Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Haifang Mao
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Jibo Liu
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Chen Chen
- Department of Food Science and Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Huaixiang Tian
- Department of Food Science and Technology, Shanghai Institute of Technology, Shanghai 201418, China
| |
Collapse
|
5
|
Liu Q, Wang M, Hou Y, Chen R, Liu H, Han T, Liu D. Deciphering the multifaceted effects of artificial sweeteners on body health and metabolic functions: a comprehensive review and future perspectives. Crit Rev Food Sci Nutr 2024:1-23. [PMID: 39368060 DOI: 10.1080/10408398.2024.2411410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2024]
Abstract
As the rates of chronic diseases such as obesity and diabetes rise worldwide, there is a growing demand for low-calorie or no-calorie sweeteners to reduce sugar intake without sacrificing the sweetness of foods and beverages. Artificial sweeteners have become indispensable as substitutes for sugar due to their high sweetening power and low impact on blood sugar levels and are used in a variety of low-calorie foods and beverages. Although artificial sweeteners offer an alternative for reducing sugar intake while maintaining sweetness, research into their long-term health effects, particularly at high doses, is ongoing, further scientific research and regulatory review are needed to clarify these potential health risks. This article reviews the latest research on the health effects of artificial sweeteners, based on recent studies, introduces the classification, performance, and safety standards for artificial sweeteners, analyses their potential harms to the nervous, immune, and circulatory systems, reproductive system, as well as their effects on gut microbiota, liver function, cancer, diabetes, and obesity. In addition, consumer perceptions of artificial sweeteners and future research directions are discussed, providing insights into current research controversies and knowledge gaps, as well as the health research and market application of artificial sweeteners.
Collapse
Affiliation(s)
- Qiang Liu
- College of Food Science and Engineering, Bohai University, Jinzhou, China
| | - Min Wang
- College of Food Science and Engineering, Bohai University, Jinzhou, China
| | - Yuting Hou
- College of Food Science and Engineering, Bohai University, Jinzhou, China
- Meat Innovation Center of Liaoning Province, Jinzhou, China
- Liaoning Kazuo Hybrid Wild Boar Science and Technology Backyard, Chaoyang, China
| | - Rui Chen
- College of Food Science and Engineering, Bohai University, Jinzhou, China
| | - Haixia Liu
- College of Food Science and Engineering, Bohai University, Jinzhou, China
| | - Tianlong Han
- College of Food Science and Engineering, Bohai University, Jinzhou, China
- Liaoning Kazuo Hybrid Wild Boar Science and Technology Backyard, Chaoyang, China
| | - Dengyong Liu
- College of Food Science and Engineering, Bohai University, Jinzhou, China
- Meat Innovation Center of Liaoning Province, Jinzhou, China
| |
Collapse
|
6
|
Kim SK, Guthrie B, Goddard WA. Ligand-Dependent and G Protein-Dependent Properties for the Sweet Taste Heterodimer, TAS1R2/1R3. J Phys Chem B 2024; 128:8927-8932. [PMID: 39231438 PMCID: PMC11421092 DOI: 10.1021/acs.jpcb.4c04610] [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: 07/09/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024]
Abstract
The heterodimeric sweet taste receptor, TAS1R2/1R3, is a class C G protein-coupled receptor (GPCR) that couples to gustducin (Gt), a G protein (GP) specifically involved in taste processing. This makes TAS1R2/1R3 a possible target for newly developing low caloric ligands that taste sweet to address obesity and diabetes. The activation of TAS1R2/1R3 involves the insertion of the GαP C-terminus of the GP into the GPCR in response to ligand binding. However, it is not known for sure whether the GP inserts into the TAS1R2 or TAS1R3 intracellular region of this GPCR dimer. Moreover, TAS1R2/1R3 can also connect to other GPs, such as Gs, Gi1, Gt3, Go, Gq, and G12. These GPs have different C-termini that may modify GPCR signaling. To understand the possible GP dependence of sweet perception, we use molecular dynamic (MD) simulations to examine the coupling of various GαP C20 termini to TAS1R2/1R3 for various steviol glycoside ligands and an artificial sweetener. Since the C20 could interact with the transmembrane domain (TMD) of either TAS1R2 (TMD2) or TAS1R3 (TMD3), we consider both cases. Without any sweetener, we find that the apo GPCR shows similar Go and Gt selectivities, while all steviol glycoside ligands increase the selectivity of Gt but decrease Go selectivity at TMD2. Interestingly, we find that high sweet rebaudioside M (RebM) and RebD ligands show better interactions of C20 at TMD3 for the Gt protein, but low sweet RebC and hydRebM ligands show better interaction of C20 at TMD2 for the Gt protein. Thus, our MD simulation suggests that TAS1R2/1R3 may couple the GP to either 1R2 or to 1R3 and that it can couple other GPs compared to Gt. This will likely lead to multimodal functions producing multiple patterns of intracellular signaling for sweet taste receptors, depending on the particular sweetener. Directing the GP to one of the other may have beneficial therapeutic outcomes.
Collapse
Affiliation(s)
- Soo-Kyung Kim
- Materials
and Process Simulation Center (139-74), California Institute of Technology, Pasadena, California 91125, United States
| | - Brian Guthrie
- Cargill
Global Core Research, Wayzata, Minnesota 55391, United States
| | - William A. Goddard
- Materials
and Process Simulation Center (139-74), California Institute of Technology, Pasadena, California 91125, United States
| |
Collapse
|
7
|
Wei F, Luo L, Tian S, Qin Y, Luo W, Zeng L. Synergistic Effect Mechanism of Binary Sweet Taste Compounds. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:20028-20036. [PMID: 39208273 DOI: 10.1021/acs.jafc.4c05004] [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: 09/04/2024]
Abstract
In our previous study, phloridzin, sucrose, l-alanine, and dulcitol presented synergistic effects in Camellia nanchuanica black tea (NCBT). This study aims to verify the synergistic effects of the aforementioned sweet taste compounds and the mechanism involved. By conducting σ-τ plot analysis, phloridzin at the recognition threshold concentration (phl) exhibited synergistic effects with different concentrations of sucrose (Lsuc-6suc). Various concentrations of sucrose, phloridzin, and their combinations were selected to investigate the impact on sweet taste receptor cells. The results revealed that sucrose/phloridzin significantly increased the calcium signal compared to phloridzin and sucrose alone, attributed to the greater stability of the sucrose/phloridzin combination when binding to Taste 1 Receptor Member 3 (TAS1R3; one subunit of sweet taste receptor proteins). Ultimately, the sweet taste signal of sucrose/phloridzin was transmitted to the brain, triggering the activation of more brain regions associated with sweet taste perception (right insular, postcentral, and amygdala).
Collapse
Affiliation(s)
- Fang Wei
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City/College of Food Science, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China
- Modern "Chuan cai Yu wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| | - Liyong Luo
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City/College of Food Science, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China
- Modern "Chuan cai Yu wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| | - Shiyi Tian
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Yumei Qin
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China
| | - Wei Luo
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City/College of Food Science, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China
- Modern "Chuan cai Yu wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| | - Liang Zeng
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City/College of Food Science, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China
- Modern "Chuan cai Yu wei" Food Industry Innovation Research Institute, Chongqing 400715, China
| |
Collapse
|
8
|
Zhang HX, Zhou HW, Liu SQ, Zheng ZF, Du ZZ. New Sweet-Tasting Gypenosides from "Jiaogulan" ( Gynostemma pentaphyllum) and Their Interactions with the Homology Model of Sweet Taste Receptors. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:18619-18629. [PMID: 39105697 DOI: 10.1021/acs.jafc.4c03566] [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: 08/07/2024]
Abstract
Gynostemma pentaphyllum has been used as an herbal tea, vegetable, and dietary supplement for hundreds of years in East Asia. The sweet variety, grown in large areas in Fujian Province, China, is an essential source of "Jiaogulan" herbal tea. However, its sweet components are unknown. To investigate the sweet constituents of Fujian "Jiaogulan" and discover new natural high-potency sweeteners, phytochemical and sensory evaluations were combined to obtain 15 saponins, of which 11 (1-11) were sweet-tasting, including 2 new ones with sweetness intensities 20-200 times higher than that of sucrose, and four (12-15) were bitter-tasting. Their structures were elucidated using spectroscopic methods (NMR, MS, IR, UV), hydrolysis, and comparison with literature data. The contents of the 15 saponins were quantitatively analyzed using UPLC-MS/MS in multiple reaction monitoring mode. The contents of 1 and 2 sweet-tasting gypenosides were 9.913 ± 1.735 and 35.852 ± 1.739 mg/kg, respectively. The content of the sweetest compound (6) was 124.969 ± 0.961 mg/kg. Additionally, compound 4 was the most abundant sweet component (422.530 ± 3.702 mg/kg). Furthermore, molecular docking results suggested interactions of sweet saponins with sweet taste receptors. In general, this study revealed the material basis of the Fujian "Jiaogulan" taste.
Collapse
Affiliation(s)
- Hong-Xia Zhang
- National-Local Joint Engineering Research Center for Highly-Efficient Utilization Technology of Forest Biomass Resources; Key Laboratory for Highly-Efficient Utilization of Forest Biomass Resources in the Southwest China, National Forestry and Grassland Administration; College of Materials and Chemical Engineering, Southwest Forestry University, Kunming 650224, China
- Department of Economic Plants and Biotechnology, Yunnan Key Laboratory for Wild Plant Resources, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Hui-Wei Zhou
- Department of Economic Plants and Biotechnology, Yunnan Key Laboratory for Wild Plant Resources, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shou-Qing Liu
- National-Local Joint Engineering Research Center for Highly-Efficient Utilization Technology of Forest Biomass Resources; Key Laboratory for Highly-Efficient Utilization of Forest Biomass Resources in the Southwest China, National Forestry and Grassland Administration; College of Materials and Chemical Engineering, Southwest Forestry University, Kunming 650224, China
| | - Zhi-Feng Zheng
- National-Local Joint Engineering Research Center for Highly-Efficient Utilization Technology of Forest Biomass Resources; Key Laboratory for Highly-Efficient Utilization of Forest Biomass Resources in the Southwest China, National Forestry and Grassland Administration; College of Materials and Chemical Engineering, Southwest Forestry University, Kunming 650224, China
- Fujian Provincial Industry Technologies Development Based for New Energy; College of Energy, Xiamen University, Xiamen 361102, China
| | - Zhi-Zhi Du
- Department of Economic Plants and Biotechnology, Yunnan Key Laboratory for Wild Plant Resources, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| |
Collapse
|
9
|
Kou X, Shi P, Gao C, Ma P, Xing H, Ke Q, Zhang D. Data-Driven Elucidation of Flavor Chemistry. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:6789-6802. [PMID: 37102791 PMCID: PMC10176570 DOI: 10.1021/acs.jafc.3c00909] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Flavor molecules are commonly used in the food industry to enhance product quality and consumer experiences but are associated with potential human health risks, highlighting the need for safer alternatives. To address these health-associated challenges and promote reasonable application, several databases for flavor molecules have been constructed. However, no existing studies have comprehensively summarized these data resources according to quality, focused fields, and potential gaps. Here, we systematically summarized 25 flavor molecule databases published within the last 20 years and revealed that data inaccessibility, untimely updates, and nonstandard flavor descriptions are the main limitations of current studies. We examined the development of computational approaches (e.g., machine learning and molecular simulation) for the identification of novel flavor molecules and discussed their major challenges regarding throughput, model interpretability, and the lack of gold-standard data sets for equitable model evaluation. Additionally, we discussed future strategies for the mining and designing of novel flavor molecules based on multi-omics and artificial intelligence to provide a new foundation for flavor science research.
Collapse
Affiliation(s)
- Xingran Kou
- Collaborative Innovation Center of Fragrance Flavour and Cosmetics, School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Peiqin Shi
- Collaborative Innovation Center of Fragrance Flavour and Cosmetics, School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Chukun Gao
- Laboratory for Physical Chemistry, ETH Zürich, 8093 Zürich, Switzerland
| | - Peihua Ma
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland 20742, United States
| | - Huadong Xing
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Qinfei Ke
- Collaborative Innovation Center of Fragrance Flavour and Cosmetics, School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Dachuan Zhang
- National Centre of Competence in Research (NCCR) Catalysis, Institute of Environmental Engineering, ETH Zürich, 8093 Zürich, Switzerland
| |
Collapse
|
10
|
Sanematsu K, Yamamoto M, Nagasato Y, Kawabata Y, Watanabe Y, Iwata S, Takai S, Toko K, Matsui T, Wada N, Shigemura N. Prediction of dynamic allostery for the transmembrane domain of the sweet taste receptor subunit, TAS1R3. Commun Biol 2023; 6:340. [PMID: 37012338 PMCID: PMC10070457 DOI: 10.1038/s42003-023-04705-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 03/14/2023] [Indexed: 04/05/2023] Open
Abstract
The sweet taste receptor plays an essential role as an energy sensor by detecting carbohydrates. However, the dynamic mechanisms of receptor activation remain unclear. Here, we describe the interactions between the transmembrane domain of the G protein-coupled sweet receptor subunit, TAS1R3, and allosteric modulators. Molecular dynamics simulations reproduced species-specific sensitivity to ligands. We found that a human-specific sweetener, cyclamate, interacted with the mouse receptor as a negative allosteric modulator. Agonist-induced allostery during receptor activation was found to destabilize the intracellular part of the receptor, which potentially interfaces with the Gα subunit, through ionic lock opening. A common human variant (R757C) of the TAS1R3 exhibited a reduced response to sweet taste, in support of our predictions. Furthermore, histidine residues in the binding site acted as pH-sensitive microswitches to modulate the sensitivity to saccharin. This study provides important insights that may facilitate the prediction of dynamic activation mechanisms for other G protein-coupled receptors.
Collapse
Affiliation(s)
- Keisuke Sanematsu
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
- Oral Health/Brain Health/Total Health Research Center, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
- Research and Development Center for Five-Sense Devices, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.
| | - Masato Yamamoto
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Department of General Dentistry, Division of Interdisciplinary Dentistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuki Nagasato
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Department of Bioresources and Biosciences, Faculty of Agriculture, Graduate School of Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Yuko Kawabata
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yu Watanabe
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Shusuke Iwata
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
- Research and Development Center for Five-Sense Devices, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Shingo Takai
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kiyoshi Toko
- Research and Development Center for Five-Sense Devices, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
- Institute for Advanced Study, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Toshiro Matsui
- Research and Development Center for Five-Sense Devices, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
- Department of Bioresources and Biosciences, Faculty of Agriculture, Graduate School of Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Naohisa Wada
- Department of General Dentistry, Division of Interdisciplinary Dentistry, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Noriatsu Shigemura
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
- Research and Development Center for Five-Sense Devices, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan.
| |
Collapse
|
11
|
Yu Y, Xu S, He R, Liang G. Application of Molecular Simulation Methods in Food Science: Status and Prospects. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:2684-2703. [PMID: 36719790 DOI: 10.1021/acs.jafc.2c06789] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Molecular simulation methods, such as molecular docking, molecular dynamic (MD) simulation, and quantum chemical (QC) calculation, have become popular as characterization and/or virtual screening tools because they can visually display interaction details that in vitro experiments can not capture and quickly screen bioactive compounds from large databases with millions of molecules. Currently, interdisciplinary research has expanded molecular simulation technology from computer aided drug design (CADD) to food science. More food scientists are supporting their hypotheses/results with this technology. To understand better the use of molecular simulation methods, it is necessary to systematically summarize the latest applications and usage trends of molecular simulation methods in the research field of food science. However, this type of review article is rare. To bridge this gap, we have comprehensively summarized the principle, combination usage, and application of molecular simulation methods in food science. We also analyzed the limitations and future trends and offered valuable strategies with the latest technologies to help food scientists use molecular simulation methods.
Collapse
Affiliation(s)
- Yuandong Yu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing400030, China
| | - Shiqi Xu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing400030, China
| | - Ran He
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing400030, China
| | - Guizhao Liang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing400030, China
| |
Collapse
|
12
|
Ramis R, Ballesteros ÓR, Muguruza-Montero A, M-Alicante S, Núñez E, Villarroel Á, Leonardo A, Bergara A. Molecular dynamics simulations of the calmodulin-induced α-helix in the SK2 calcium-gated potassium ion channel. J Biol Chem 2022; 299:102850. [PMID: 36587765 PMCID: PMC9874072 DOI: 10.1016/j.jbc.2022.102850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 12/30/2022] Open
Abstract
The family of small-conductance Ca2+-activated potassium ion channels (SK channels) is composed of four members (SK1, SK2, SK3, and SK4) involved in neuron-firing regulation. The gating of these channels depends on the intracellular Ca2+ concentration, and their sensitivity to this ion is provided by calmodulin (CaM). This protein binds to a specific region in SK channels known as the calmodulin-binding domain (CaMBD), an event which is essential for their gating. While CaMBDs are typically disordered in the absence of CaM, the SK2 channel subtype displays a small prefolded α-helical region in its CaMBD even if CaM is not present. This small helix is known to turn into a full α-helix upon CaM binding, although the molecular-level details for this conversion are not fully understood yet. In this work, we offer new insights on this physiologically relevant process by means of enhanced sampling, atomistic Hamiltonian replica exchange molecular dynamics simulations, providing a more detailed understanding of CaM binding to this target. Our results show that CaM is necessary for inducing a full α-helix along the SK2 CaMBD through hydrophobic interactions with V426 and L427. However, it is also necessary that W431 does not compete for these interactions; the role of the small prefolded α-helix in the SK2 CaMBD would be to stabilize W431 so that this is the case. In conclusion, our findings provide further insight into a key interaction between CaM and SK channels that is important for channel sensitivity to Ca2+.
Collapse
Affiliation(s)
- Rafael Ramis
- Donostia International Physics Center, Donostia, Spain; Departamento de Física, Universidad del País Vasco, UPV/EHU, Leioa, Spain.
| | - Óscar R. Ballesteros
- Departamento de Física, Universidad del País Vasco, UPV/EHU, Leioa, Spain,Centro de Física de Materiales CFM, CSIC-UPV/EHU, Donostia, Spain
| | | | - Sara M-Alicante
- Departamento de Física, Universidad del País Vasco, UPV/EHU, Leioa, Spain,Instituto Biofisika, CSIC-UPV/EHU, Leioa, Spain
| | - Eider Núñez
- Departamento de Física, Universidad del País Vasco, UPV/EHU, Leioa, Spain,Instituto Biofisika, CSIC-UPV/EHU, Leioa, Spain
| | | | - Aritz Leonardo
- Donostia International Physics Center, Donostia, Spain,Departamento de Física, Universidad del País Vasco, UPV/EHU, Leioa, Spain
| | - Aitor Bergara
- Donostia International Physics Center, Donostia, Spain,Departamento de Física, Universidad del País Vasco, UPV/EHU, Leioa, Spain,Centro de Física de Materiales CFM, CSIC-UPV/EHU, Donostia, Spain
| |
Collapse
|
13
|
Laffitte A, Belloir C, Neiers F, Briand L. Functional Characterization of the Venus Flytrap Domain of the Human TAS1R2 Sweet Taste Receptor. Int J Mol Sci 2022; 23:ijms23169216. [PMID: 36012481 PMCID: PMC9409066 DOI: 10.3390/ijms23169216] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
The human sweet taste receptor is a heterodimeric receptor composed of two distinct G-protein-coupled receptors (GPCRs), TAS1R2 and TAS1R3. The TAS1R2 and TAS1R3 subunits are members of a small family of class C GPCRs whose members share the same architecture, comprising a Venus Flytrap (VFT) module linked to the seven transmembrane domains (TMDs) by a short cysteine-rich region (CRR). The VFT module of TAS1R2 contains the primary binding site for most of the sweet-tasting compounds, including natural sugars and artificial and natural sweeteners. However, cellular assays, molecular docking and site-directed mutagenesis studies have revealed that the VFT, CRR and TMD of TAS1R3 interact with some sweeteners, including the sweet-tasting protein brazzein. The aim of this study was to better understand the contribution of TAS1R2-VFT in the binding of sweet stimuli. To achieve this, we heterologously expressed human TAS1R2-VFT (hTAS1R2-VFT) in Escherichia coli. Circular dichroism spectroscopic studies revealed that hTAS1R2-VFT was properly folded with evidence of secondary structures. Using size-exclusion chromatography coupled with light scattering, we found that hTAS1R2-VFT behaves as a monomer. Ligand binding quantified by intrinsic tryptophan fluorescence showed that hTAS1R2-VFT is capable of binding sweet stimuli with Kd values, in agreement with physiological detection. Furthermore, we investigated whether the impact of point mutations, already shown to have deleterious effects on cellular assays, could impact the ability of hTAS1R2-VFT to bind sweet ligands. As expected, the ligand affinities of hTAS1R2-VFT were drastically reduced through the introduction of single amino acid substitutions (D278A and E382A) known to abolish the response of the full-length TAS1R2/TAS1R3 receptor. This study demonstrates the feasibility of producing milligram quantities of hTAS1R2-VFT to further characterize the mechanism of binding interaction and perform structural studies.
Collapse
|
14
|
Current Advances and Future Aspects of Sweetener Synergy: Properties, Evaluation Methods and Molecular Mechanisms. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12105096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Sweetener synergy is the phenomenon in which certain combinations of sweeteners work more effectively than the theoretical sum of the effects of each components. It provides benefits in reducing sweetener dosages and improving their sweetness. Many mixtures of sweeteners with synergistic effects have been reported up to now. Both artificial high-intensity sweeteners and natural sweeteners are popularly used in sweetener mixtures for synergism, although the former seem to display more potential to exhibit synergy than the latter. Furthermore, several evaluation methods to investigate sweetener synergy have been applied, which could lead to discrepancies in results. Moreover, structurally dissimilar sweeteners could cooperatively bind at the different sites in the sweet taste receptor T1R2/T1R3 to activate the receptor, and their hydration characters/packing characteristics in solvents could affect their interaction with the receptor, providing the preliminary explanations for the molecular basis of sweetener synergy. In this article, we firstly present a systematic review, analysis and comment on the properties, evaluation methods and molecular mechanisms of sweetener synergy. Secondly, challenges of sweetener synergy in both theory and practice and possible strategies to overcome these limitations are comprehensively discussed. Finally, future perspectives for this important performance in human sweet taste perception are proposed.
Collapse
|