1
|
Gong SM, Jiang Y, Xue YB, Peng YY, Qian CY, Zhang Y, Zhou R, Huang L. The odorant (R)-(-)-carvone promotes glucose-stimulated insulin secretion via the olfactory receptor Olfr1259 in pancreatic β-TC6 cells. Arch Biochem Biophys 2025; 768:110404. [PMID: 40157529 DOI: 10.1016/j.abb.2025.110404] [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: 01/19/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Olfactory receptors (ORs) make up the largest subfamily of G protein-coupled receptors that are expressed in olfactory sensory neurons in the nasal cavity and recognize an enormous number of odorants from the external environment. These receptors, however, have also been found in many other tissues including pancreas, liver, and adipose tissue, in which they seem to play important but different roles. Yet, the exact functions of ORs in these extra-nasal tissues are not well understood. Here, we report that (R)-(-)-carvone and a few other odorants were able to evoke calcium responses in mouse pancreatic β-TC6 cells. Furthermore, (R)-(-)-carvone potentiated cytoplasmic cAMP accumulation and glucose-stimulated insulin secretion (GSIS). More importantly, GPCR signaling pathway components adenylyl cyclase, phospholipase C, and inositol triphosphate receptor were involved in (R)-(-)-carvone-induced signal transduction. By reanalyzing the available β-TC6 cells' RNAseq dataset, we identified several candidate ORs for (R)-(-)-carvone. Further analyses with molecular docking and molecular dynamics simulations indicated that (R)-(-)-carvone bound to the odorant-binding pocket of the olfactory receptor Olfr1259 while knockdown of Olfr1259 expression in β-TC6 cells with siRNA significantly reduced the stimulatory effects of (R)-(-)-carvone on cytoplasmic Ca2+ and cAMP levels, and insulin secretion. Together, these results indicated that Olfr1259 is the receptor for (R)-(-)-carvone in β-TC6 cells. Therefore, our study highlighted the important roles of (R)-(-)-carvone and its receptor Olfr1259 in initiating calcium signaling, inducing intracellular cAMP accumulation, and enhancing GSIS in pancreatic β cells, demonstrating that Olfr1259 may be a new therapeutic target for regulating glucose metabolism and for treating diabetes.
Collapse
Affiliation(s)
- Shi-Meng Gong
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yangwei Jiang
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yan-Bo Xue
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yuan-Yuan Peng
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Chun-Yan Qian
- Linping Branch of the Second Affiliated Hospital, Zhejiang University of School of Medicine, Hangzhou, Zhejiang, 311100, China
| | - Yue Zhang
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Ruhong Zhou
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China; The First Affiliated Hospital, Zhejiang University of School of Medicine, Hangzhou, Zhejiang, 310058, China.
| | - Liquan Huang
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
2
|
Niu Y, Wei J, Zhu J, Feng H, Ren Y, Guo Z, Zhang J, Zhou R, She Y, Wang Z, Xiao Z. Study on the mechanism of glucose-lowering and sweetening of key sweet aroma compounds in sweet orange. Food Res Int 2024; 196:114874. [PMID: 39614462 DOI: 10.1016/j.foodres.2024.114874] [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: 05/10/2024] [Revised: 07/25/2024] [Accepted: 08/05/2024] [Indexed: 12/01/2024]
Abstract
Rising living standards heighten the demand for healthier sugar-reduced foods. This study used Headspace Solid-Phase Microextraction (HS-SPME) and Gas Chromatography-Olfactometry-Mass Spectrometry (GC/O-AT) to analyze volatile components in sweet orange juice, identifying 12 key sweet aroma compounds. Sensory and electronic tongue evaluations indicated that seven sweetness-related odor substances- (E)-citral, (E)-β-farnesene, β-myrcene, tallo-ocimene, nonanal, citronellyl formate, and tallo-ocimene-significantly enhanced the sweetness of a 5 % sucrose solution. In contrast, while nonanol was found to have no sugar-reducing sweetness-enhancing effect. Furthermore, molecular docking analysis was employed to examine the regions, binding energies, and interaction forces between eight sweet and fragrant aroma compounds from sweet orange and the T1R2-T1R3 sweet taste receptor-sucrose ternary system. The average binding energies with the receptor were -3.2 kcal/mol, -1.2 kcal/mol, -3.0 kcal/mol, -1.6 kcal/mol, -5.9 kcal/mol, -5.8 kcal/mol, -3.6 kcal/mol, and -6.0 kcal/mol, respectively. However, it should be noted that binding energy alone is not the sole criterion for judging the sweetening effect. Molecular dynamics (MD) results further demonstrated that the stability of the binding between sucrose and the sweet taste receptor was improved under the influence of (E)-citral, with the interaction between the two relying on hydrogen bonds, water bridges, and hydrophobic forces. This provides a theoretical basis for validating the sweetness-enhancing effects of aroma substances and insights into novel sweetener development.
Collapse
Affiliation(s)
- YunWei Niu
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Jie Wei
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - JianCai Zhu
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - HaoHua Feng
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - YuKun Ren
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Zhimin Guo
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Jing Zhang
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - Rujuan Zhou
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China
| | - YuanBin She
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Zhaogai Wang
- Agricultural Products Processing Center, Henan Academy of Agricultural Sciences, Zhengzhou, China
| | - ZuoBing Xiao
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai 201418, China; School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China; Agricultural Products Processing Center, Henan Academy of Agricultural Sciences, Zhengzhou, China.
| |
Collapse
|
3
|
Maaroufi H. Novel gurmarin-like peptides from Gymnema sylvestre and their interactions with the sweet taste receptor T1R2/T1R3. Chem Senses 2024; 49:bjae018. [PMID: 38695158 PMCID: PMC11103048 DOI: 10.1093/chemse/bjae018] [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: 05/21/2024] Open
Abstract
Gymnema sylvestre (GS) is a traditional medicinal plant known for its hypoglycemic and hypolipidemic effects. Gurmarin (hereafter Gur-1) is the only known active peptide in GS. Gur-1 has a suppressive sweet taste effect in rodents but no or only a very weak effect in humans. Here, 8 gurmarin-like peptides (Gur-2 to Gur-9) and their isoforms are reported in the GS transcriptome. The molecular mechanism of sweet taste suppression by Gur-1 is still largely unknown. Therefore, the complete architecture of human and mouse sweet taste receptors T1R2/T1R3 and their interaction with Gur-1 to Gur-9 were predicted by AlphaFold-Multimer (AF-M) and validated. Only Gur-1 and Gur-2 interact with the T1R2/T1R3 receptor. Indeed, Gur-1 and Gur-2 bind to the region of the cysteine-rich domain (CRD) and the transmembrane domain (TMD) of the mouse T1R2 subunit. In contrast, only Gur-2 binds to the TMD of the human T1R2 subunit. This result suggests that Gur-2 may have a suppressive sweet taste effect in humans. Furthermore, AF-M predicted that Gα-gustducin, a protein involved in sweet taste transduction, interacts with the intracellular domain of the T1R2 subunit. These results highlight an unexpected diversity of gurmarin-like peptides in GS and provide the complete predicted architecture of the human and mouse sweet taste receptor with the putative binding sites of Gur-1, Gur-2, and Gα-gustducin. In addition, gurmarin-like peptides may serve as promising drug scaffolds for the development of antidiabetic molecules.
Collapse
Affiliation(s)
- Halim Maaroufi
- Institut de biologie intégrative et des systèmes (IBIS), Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|
4
|
John YJ, Caldwell L, McCoy DE, Braganza O. Dead rats, dopamine, performance metrics, and peacock tails: Proxy failure is an inherent risk in goal-oriented systems. Behav Brain Sci 2023; 47:e67. [PMID: 37357710 DOI: 10.1017/s0140525x23002753] [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] [Indexed: 06/27/2023]
Abstract
When a measure becomes a target, it ceases to be a good measure. For example, when standardized test scores in education become targets, teachers may start "teaching to the test," leading to breakdown of the relationship between the measure - test performance - and the underlying goal - quality education. Similar phenomena have been named and described across a broad range of contexts, such as economics, academia, machine learning, and ecology. Yet it remains unclear whether these phenomena bear only superficial similarities, or if they derive from some fundamental unifying mechanism. Here, we propose such a unifying mechanism, which we label proxy failure. We first review illustrative examples and their labels, such as the "cobra effect," "Goodhart's law," and "Campbell's law." Second, we identify central prerequisites and constraints of proxy failure, noting that it is often only a partial failure or divergence. We argue that whenever incentivization or selection is based on an imperfect proxy measure of the underlying goal, a pressure arises that tends to make the proxy a worse approximation of the goal. Third, we develop this perspective for three concrete contexts, namely neuroscience, economics, and ecology, highlighting similarities and differences. Fourth, we outline consequences of proxy failure, suggesting it is key to understanding the structure and evolution of goal-oriented systems. Our account draws on a broad range of disciplines, but we can only scratch the surface within each. We thus hope the present account elicits a collaborative enterprise, entailing both critical discussion as well as extensions in contexts we have missed.
Collapse
Affiliation(s)
- Yohan J John
- Neural Systems Laboratory, Department of Health and Rehabilitation Sciences, Boston University, Boston, MA, USA
| | | | - Dakota E McCoy
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Hopkins Marine Station, Stanford University, Pacific Grove, CA, USA
- Department of Biology, Duke University, Durham, NC, USA
| | - Oliver Braganza
- Institute for Experimental Epileptology and Cognition Research, University of Bonn, Bonn, Germany
- Institute for Socioeconomics, University of Duisburg-Essen, Duisburg, Germany
| |
Collapse
|
5
|
Evangelista-Falcón W, Denhez C, Baena-Moncada A, Ponce-Vargas M. Revisiting the Sweet Taste Receptor T1R2-T1R3 through Molecular Dynamics Simulations Coupled with a Noncovalent Interactions Analysis. J Phys Chem B 2023; 127:1110-1119. [PMID: 36705604 DOI: 10.1021/acs.jpcb.2c07180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
It is nowadays widely accepted that sweet taste perception is elicited by the activation of the heterodimeric complex T1R2-T1R3, customarily known as sweet taste receptor (STR). However, the interplay between STR and sweeteners has not yet been fully clarified. Here through a methodology coupling molecular dynamics and the independent gradient model (igm) approach we determine the main interacting signatures of the closed (active) conformation of the T1R2 Venus flytrap domain (VFD) toward aspartame. The igm methodology provides a rapid and reliable quantification of noncovalent interactions through a score (Δginter) based on the attenuation of the electronic density gradient when two molecular fragments approach each other. Herein, this approach is coupled to a 100 ns molecular dynamics simulation (MD-igm) to explore the ligand-cavity contacts on a per-residue basis as well as a series of key inter-residue interactions that stabilize the closed form of VFD. We also apply an atomic decomposition scheme of noncovalent interactions to quantify the contribution of the ligand segments to the noncovalent interplay. Finally, a series of structural modification on aspartame are conducted in order to obtain guidelines for the rational design of novel sweeteners. Given that innovative methodologies to reliably quantify the extent of ligand-protein coupling are strongly demanded, this approach combining a noncovalent analysis and MD simulations represents a valuable contribution, that can be easily applied to other relevant biomolecular systems.
Collapse
Affiliation(s)
- Wilfredo Evangelista-Falcón
- Laboratory of Biomolecules, Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Lima15023, Perú
| | - Clément Denhez
- Institut de Chimie Moléculaire de Reims UMR CNRS 7312, Université de Reims Champagne-Ardenne, Moulin de la Housse 51687, ReimsCedex 02 BP39, France
| | - Angélica Baena-Moncada
- Laboratorio de Investigación de Electroquímica Aplicada, Facultad de Ciencias de la Universidad Nacional de Ingeniería, Av. Túpac Amaru 210, Rímac, Lima31-139, Perú
| | - Miguel Ponce-Vargas
- Institut de Chimie Moléculaire de Reims UMR CNRS 7312, Université de Reims Champagne-Ardenne, Moulin de la Housse 51687, ReimsCedex 02 BP39, France
| |
Collapse
|
6
|
Ponnusamy V, Subramanian G, Muthuswamy K, Shanmugamprema D, Krishnan V, Velusamy T, Subramaniam S. Genetic variation in sweet taste receptors and a mechanistic perspective on sweet and fat taste sensation in the context of obesity. Obes Rev 2022; 23:e13512. [PMID: 36282093 DOI: 10.1111/obr.13512] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/14/2022] [Accepted: 10/05/2022] [Indexed: 11/30/2022]
Abstract
Taste sensation enables humans to make nutritionally important decisions such as food preference and consumption. It functions as deterministic factors for unpropitious eating behavior, leading to overweight and obesity. The hedonistic feeling on consumption of fat and sugar-rich meals, in particular, has a negative influence on health. In addition, impairment in the taste receptors alters the downstream signaling of taste transduction pathway. Hence, genetic polymorphism in typical taste receptors is a predictor of taste sensitivity variance across individuals. The present review summarizes the effect of a single nucleotide polymorphism (SNP) in sweet taste receptors (T1R2/T1R3) on taste perception among individuals of various body mass index (BMI). Furthermore, in the context of obesity, we discussed the possibility of crosstalk between fat and sweet receptors as well as taste dysfunction in diseased individuals. In overall, a greater understanding of the physiological relationship between taste receptors, altered taste sensitivity, and genetic polymorphisms should lead to more effective obesity prevention approaches.
Collapse
Affiliation(s)
- Vinithra Ponnusamy
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamilnadu, India, 641046
| | - Gowtham Subramanian
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamilnadu, India, 641046
| | - Karthi Muthuswamy
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamilnadu, India, 641046
| | - Deepankumar Shanmugamprema
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamilnadu, India, 641046
| | - Vasanth Krishnan
- Molecular Biology Laboratory, Department of Botany, School of Life Sciences, Bharathiar University, Coimbatore, Tamilnadu, India, 641046
| | - Thirunavukkarasu Velusamy
- Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, Tamilnadu, India, 641046
| | - Selvakumar Subramaniam
- Molecular Physiology Laboratory, Department of Biochemistry, Bharathiar University, Coimbatore, Tamilnadu, India, 641046
| |
Collapse
|
7
|
Zhao Y, Dávila EM, Li X, Tang B, Rabinowitsch AI, Perez-Aguilar JM, Blobel CP. Identification of Molecular Determinants in iRhoms1 and 2 That Contribute to the Substrate Selectivity of Stimulated ADAM17. Int J Mol Sci 2022; 23:12796. [PMID: 36361585 PMCID: PMC9654401 DOI: 10.3390/ijms232112796] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/03/2022] [Accepted: 10/19/2022] [Indexed: 09/29/2023] Open
Abstract
The metalloprotease ADAM17 is a key regulator of the TNFα, IL-6R and EGFR signaling pathways. The maturation and function of ADAM17 is controlled by the seven-membrane-spanning proteins iRhoms1 and 2. The functional properties of the ADAM17/iRhom1 and ADAM17/iRhom2 complexes differ, in that stimulated shedding of most ADAM17 substrates tested to date can be supported by iRhom2, whereas iRhom1 can only support stimulated shedding of very few ADAM17 substrates, such as TGFα. The first transmembrane domain (TMD1) of iRhom2 and the sole TMD of ADAM17 are important for the stimulated shedding of ADAM17 substrates by iRhom2. However, little is currently known about how the iRhoms interact with different substrates to control their stimulated shedding by ADAM17. To provide new insights into this topic, we tested how various chimeras between iRhom1 and iRhom2 affect the stimulated processing of the EGFR-ligands TGFα (iRhom1- or 2-dependent) and EREG (iRhom2-selective) by ADAM17. This uncovered an important role for the TMD7 of the iRhoms in determining their substrate selectivity. Computational methods utilized to characterize the iRhom1/2/substrate interactions suggest that the substrate selectivity is determined, at least in part, by a distinct accessibility of the substrate cleavage site to stimulated ADAM17. These studies not only provide new insights into why the substrate selectivity of stimulated iRhom2/ADAM17 differs from that of iRhom1/ADAM17, but also suggest new approaches for targeting the release of specific ADAM17 substrates.
Collapse
Affiliation(s)
- Yi Zhao
- Department of Biochemistry, Cellular and Molecular Biology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Eliud Morales Dávila
- School of Chemical Sciences, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla 72570, Mexico
| | - Xue Li
- Department of Biochemistry, Cellular and Molecular Biology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Beiyu Tang
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ariana I. Rabinowitsch
- Department of Biochemistry, Cellular and Molecular Biology, Weill Cornell Medicine, New York, NY 10021, USA
- Weill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program, New York, NY 10021, USA
| | - Jose Manuel Perez-Aguilar
- School of Chemical Sciences, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla 72570, Mexico
| | - Carl P. Blobel
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY 10021, USA
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY 10021, USA
| |
Collapse
|
8
|
Dávila EM, Patricio F, Rebolledo-Bustillo M, Garcia-Gomez D, Hernandez JCG, Sanchez-Gaytan BL, Limón ID, Perez-Aguilar JM. Interacting binding insights and conformational consequences of the differential activity of cannabidiol with two endocannabinoid-activated G-protein-coupled receptors. Front Pharmacol 2022; 13:945935. [PMID: 36016551 PMCID: PMC9395587 DOI: 10.3389/fphar.2022.945935] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/12/2022] [Indexed: 11/25/2022] Open
Abstract
Cannabidiol (CBD), the major non-psychoactive phytocannabinoid present in the plant Cannabis sativa, has displayed beneficial pharmacological effects in the treatment of several neurological disorders including, epilepsy, Parkinson’s disease, and Alzheimer’s disease. In particular, CBD is able to modulate different receptors in the endocannabinoid system, some of which belong to the family of G-protein-coupled receptors (GPCRs). Notably, while CBD is able to antagonize some GPCRs in the endocannabinoid system, it also seems to activate others. The details of this dual contrasting functional feature of CBD, that is, displaying antagonistic and (possible) agonistic ligand properties in related receptors, remain unknown. Here, using computational methods, we investigate the interacting determinants of CBD in two closely related endocannabinoid-activated GPCRs, the G-protein-coupled receptor 55 (GPR55) and the cannabinoid type 1 receptor (CB1). While in the former, CBD has been demonstrated to function as an antagonist, the way by which CBD modulates the CB1 receptor remains unclear. Namely, CBD has been suggested to directly trigger receptor’s activation, stabilize CB1 inactive conformations or function as an allosteric modulator. From microsecond-length unbiased molecular dynamics simulations, we found that the presence of the CBD ligand in the GPR55 receptor elicit conformational changes associated with antagonist-bound GPCRs. In contrast, when the GPR55 receptor is simulated in complex with the selective agonist ML186, agonist-like conformations are sampled. These results are in agreement with the proposed modulatory function of each ligand, showing that the computational techniques utilized to characterize the GPR55 complexes correctly differentiate the agonist-bound and antagonist-bound systems. Prompted by these results, we investigated the role of the CBD compound on the CB1 receptor using similar computational approaches. The all-atom MD simulations reveal that CBD induces conformational changes linked with agonist-bound GPCRs. To contextualize the results we looked into the CB1 receptor in complex with a well-established antagonist. In contrast to the CBD/CB1 complex, when the CB1 receptor is simulated in complex with the ligand antagonist AM251, inactive conformations are explored, showing that the computational techniques utilized to characterize the CB1 complexes correctly differentiate the agonist-bound and antagonist-bound systems. In addition, our results suggest a previously unknown sodium-binding site located in the extracellular domain of the CB1 receptor. From our detailed characterization, we found particular interacting loci in the binding sites of the GPR55 and the CB1 receptors that seem to be responsible for the differential functional features of CBD. Our work will pave the way for understanding the CBD pharmacology at a molecular level and aid in harnessing its potential therapeutic use.
Collapse
Affiliation(s)
- Eliud Morales Dávila
- School of Chemical Sciences, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla, Mexico
| | - Felipe Patricio
- Neuropharmacology Laboratory, School of Chemical Sciences, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla, Mexico
| | - Mariana Rebolledo-Bustillo
- School of Chemical Sciences, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla, Mexico
| | - David Garcia-Gomez
- School of Chemical Sciences, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla, Mexico
| | - Juan Carlos Garcia Hernandez
- School of Chemical Sciences, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla, Mexico
| | - Brenda L Sanchez-Gaytan
- Chemistry Center, Science Institute, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla, Mexico
| | - Ilhuicamina Daniel Limón
- Neuropharmacology Laboratory, School of Chemical Sciences, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla, Mexico
| | - Jose Manuel Perez-Aguilar
- School of Chemical Sciences, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla, Mexico
| |
Collapse
|
9
|
Xiu H, Liu Y, Yang H, Ren H, Luo B, Wang Z, Shao H, Wang F, Zhang J, Wang Y. Identification of novel umami molecules via QSAR models and molecular docking. Food Funct 2022; 13:7529-7539. [PMID: 35765918 DOI: 10.1039/d2fo00544a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Umami substances can increase the overall taste of food and bring pleasure to people. However, it is still challenging to identify the umami molecules through virtual screening due to the crystal structure of the umami receptor being undefined. Herein, based on the hypothesis that the molecules with bitter and sweet taste characteristics may be umami molecules, this study proposed an in silico method to identify novel umami-tasting molecules in batch from SWEET-DB and BitterDB databases via the QSAR models, PCA, molecular docking and electronic tongue analysis. In total, 169 potential umami molecules were identified through QSAR modeling, PCA, and molecular docking. Of the 169 molecules, 18 were randomly selected, and all were identified as umami molecules via electronic tongue analysis. Among the 18 chosen molecules, 10 molecules could be traced back to their concentration range in food, and finally, 8 molecules were predicted to be nontoxic. This work provides a simple and efficient strategy to identify novel umami molecules, holding an excellent promise for demonstrating the crystal structure of umami receptors and taste-sensing mechanisms. Furthermore, this study opens the possibility for the practical application of new umami molecules in food.
Collapse
Affiliation(s)
- Hongxia Xiu
- Institute of Agro-Products Processing Science and Technology, Chinese Academy of Agricultural Sciences/Key Laboratory of Agro-Products Processing, Ministry of Agriculture, Beijing 100193, PR China. .,CangZhou Academy of Agriculture and Forestry Sciences, Cangzhou, 061001, PR China.
| | - Yajie Liu
- Department of Food Science, Northeast Agricultural University, Harbin, 150030, PR China
| | - Huihui Yang
- Department of Food Science, Northeast Agricultural University, Harbin, 150030, PR China
| | - Haibin Ren
- Department of Food Science, Northeast Agricultural University, Harbin, 150030, PR China
| | - Bowen Luo
- Department of Food Science, Northeast Agricultural University, Harbin, 150030, PR China
| | - Zhipeng Wang
- Department of Food Science, Northeast Agricultural University, Harbin, 150030, PR China
| | - Hong Shao
- Department of Food Science, Northeast Agricultural University, Harbin, 150030, PR China.,Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, 150030, PR China
| | - Fengzhong Wang
- Institute of Agro-Products Processing Science and Technology, Chinese Academy of Agricultural Sciences/Key Laboratory of Agro-Products Processing, Ministry of Agriculture, Beijing 100193, PR China.
| | - Jingjian Zhang
- CangZhou Academy of Agriculture and Forestry Sciences, Cangzhou, 061001, PR China.
| | - Yutang Wang
- Institute of Agro-Products Processing Science and Technology, Chinese Academy of Agricultural Sciences/Key Laboratory of Agro-Products Processing, Ministry of Agriculture, Beijing 100193, PR China. .,Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, 150030, PR China
| |
Collapse
|
10
|
Miao Y, Ni H, Zhang X, Zhi F, Long X, Yang X, He X, Zhang L. Investigating mechanism of sweetness intensity differences through dynamic analysis of sweetener-T1R2-membrane systems. Food Chem 2021; 374:131807. [PMID: 34915374 DOI: 10.1016/j.foodchem.2021.131807] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 11/19/2021] [Accepted: 12/04/2021] [Indexed: 11/15/2022]
Abstract
Knowing the mechanism of action of sweet taste receptors is important for the design of new, healthy sweeteners. However, little is known about the structures and recognition mechanisms of these receptors. 28 sweeteners were assessed by molecular docking, and 8 typical sweeteners were chosen to construct sweetener-T1R2-membrane systems to analyze interactions between receptor and sweeteners. Natural sweeteners with low-intensity sweetness, such as fructose and xylitol, were released from the Venus flytrap domain at ∼30 ns, with displacements greater than 50 Å. In contrast, artificial neotame and advantame bound stably to the receptor, within 5 Å. Van der Waals interactions were significant in high-intensity sweetener systems, imparting an energy difference of over 15 kcal/mol between neotame (artificial sweetener) and fructose (natural). These results provide a deeper understanding of the mechanisms of sweetener function and offer a new direction for the design of sweeteners.
Collapse
Affiliation(s)
- Yulu Miao
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Hui Ni
- College of Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Xingyi Zhang
- College of Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Fengdong Zhi
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Xiang Long
- College of Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Xuepeng Yang
- School of Food and Biological Engineering, Zhengzhou University of Light Industry, Zhengzhou 450002, China
| | - Xiao He
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China; NYU-ECNU Center for Computational Chemistry at NYU Shanghai, Shanghai 200062, China.
| | - Lujia Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics & New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China; NYU-ECNU Center for Computational Chemistry at NYU Shanghai, Shanghai 200062, China.
| |
Collapse
|
11
|
Róg T, Girych M, Bunker A. Mechanistic Understanding from Molecular Dynamics in Pharmaceutical Research 2: Lipid Membrane in Drug Design. Pharmaceuticals (Basel) 2021; 14:1062. [PMID: 34681286 PMCID: PMC8537670 DOI: 10.3390/ph14101062] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 11/17/2022] Open
Abstract
We review the use of molecular dynamics (MD) simulation as a drug design tool in the context of the role that the lipid membrane can play in drug action, i.e., the interaction between candidate drug molecules and lipid membranes. In the standard "lock and key" paradigm, only the interaction between the drug and a specific active site of a specific protein is considered; the environment in which the drug acts is, from a biophysical perspective, far more complex than this. The possible mechanisms though which a drug can be designed to tinker with physiological processes are significantly broader than merely fitting to a single active site of a single protein. In this paper, we focus on the role of the lipid membrane, arguably the most important element outside the proteins themselves, as a case study. We discuss work that has been carried out, using MD simulation, concerning the transfection of drugs through membranes that act as biological barriers in the path of the drugs, the behavior of drug molecules within membranes, how their collective behavior can affect the structure and properties of the membrane and, finally, the role lipid membranes, to which the vast majority of drug target proteins are associated, can play in mediating the interaction between drug and target protein. This review paper is the second in a two-part series covering MD simulation as a tool in pharmaceutical research; both are designed as pedagogical review papers aimed at both pharmaceutical scientists interested in exploring how the tool of MD simulation can be applied to their research and computational scientists interested in exploring the possibility of a pharmaceutical context for their research.
Collapse
Affiliation(s)
- Tomasz Róg
- Department of Physics, University of Helsinki, 00014 Helsinki, Finland;
| | - Mykhailo Girych
- Department of Physics, University of Helsinki, 00014 Helsinki, Finland;
| | - Alex Bunker
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland;
| |
Collapse
|
12
|
On the human taste perception: Molecular-level understanding empowered by computational methods. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.07.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
13
|
Zhang L, Domeniconi G, Yang CC, Kang SG, Zhou R, Cong G. CASTELO: clustered atom subtypes aided lead optimization-a combined machine learning and molecular modeling method. BMC Bioinformatics 2021; 22:338. [PMID: 34157976 PMCID: PMC8218488 DOI: 10.1186/s12859-021-04214-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/18/2021] [Indexed: 01/18/2023] Open
Abstract
Background Drug discovery is a multi-stage process that comprises two costly major steps: pre-clinical research and clinical trials. Among its stages, lead optimization easily consumes more than half of the pre-clinical budget. We propose a combined machine learning and molecular modeling approach that partially automates lead optimization workflow in silico, providing suggestions for modification hot spots. Results The initial data collection is achieved with physics-based molecular dynamics simulation. Contact matrices are calculated as the preliminary features extracted from the simulations. To take advantage of the temporal information from the simulations, we enhanced contact matrices data with temporal dynamism representation, which are then modeled with unsupervised convolutional variational autoencoder (CVAE). Finally, conventional and CVAE-based clustering methods are compared with metrics to rank the submolecular structures and propose potential candidates for lead optimization. Conclusion With no need for extensive structure-activity data, our method provides new hints for drug modification hotspots which can be used to improve drug potency and reduce the lead optimization time. It can potentially become a valuable tool for medicinal chemists. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-021-04214-4.
Collapse
Affiliation(s)
- Leili Zhang
- IBM Thomas J. Watson Research Center, 1101 Kitchawan Rd, 10598, Yorktown Heights, NY, USA.
| | - Giacomo Domeniconi
- IBM Thomas J. Watson Research Center, 1101 Kitchawan Rd, 10598, Yorktown Heights, NY, USA.
| | - Chih-Chieh Yang
- IBM Thomas J. Watson Research Center, 1101 Kitchawan Rd, 10598, Yorktown Heights, NY, USA
| | - Seung-Gu Kang
- IBM Thomas J. Watson Research Center, 1101 Kitchawan Rd, 10598, Yorktown Heights, NY, USA
| | - Ruhong Zhou
- ZheJiang University, 688 Yuhangtang Road, Hangzhou, 310027, China
| | - Guojing Cong
- Oak Ridge national laboratory, 1 Bethel Valley Rd, 37830, Oak Ridge, TN, USA
| |
Collapse
|
14
|
Rangel-Galván M, Rangel A, Romero-Méndez C, Dávila EM, Castro ME, Caballero NA, Meléndez Bustamante FJ, Sanchez-Gaytan BL, Meza U, Perez-Aguilar JM. Inhibitory Mechanism of the Isoflavone Derivative Genistein in the Human Ca V3.3 Channel. ACS Chem Neurosci 2021; 12:651-659. [PMID: 33507062 DOI: 10.1021/acschemneuro.0c00684] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Regulation of cellular excitability and oscillatory behavior of resting membrane potential in nerve cells are largely mediated by the low-voltage activated T-type calcium channels. This calcium channel family is constituted by three isoforms, namely, CaV3.1, CaV3.2, and CaV3.3, that are largely distributed in the nervous system and other parts of the body. Dysfunction of T-type calcium channels is associated with a wide range of pathophysiologies including epilepsy, neuropathic pain, cardiac problems, and major depressive disorders. Due to their pharmacological relevance, finding molecular agents able to modulate the channel's function may provide therapeutic means to ameliorate their related disorders. Here we used electrophysiological experiments to show that genistein, a canonical tyrosine kinase inhibitor, reduces the activity of the human CaV3.3 channel in a concentration-dependent manner. The inhibitory effect of genistein is independent of tyrosine kinase modulation and does not affect the voltage-dependent gating of the channel. Subsequently, we used computational methods to identify plausible molecular poses for the interaction of genistein and the CaV3.3 channel. Starting from different molecular poses, we carried out all-atom molecular dynamics (MD) simulations to identify the interacting determinants for the CaV3.3/genistein complex formation. Our extensive (microsecond-length) simulations suggest specific binding interactions that seem to stabilize the protein/inhibitor complex. Furthermore, our results from the unbiased MD simulations are in good agreement with the recently solved cryoelectron microscopy structure of the CaV3.1/Z944 complex in terms of both the location of the ligand binding site and the role of several equivalent amino acid residues. Proposed interacting complex loci were subsequently tested and corroborated by electrophysiological experiments using another naturally occurring isoflavone derivative, daidzein. Thus, by using a combination of in vitro and in silico techniques, we have identified interacting determinants relevant to the CaV3.3/genistein complex formation and propose that genistein directly blocks the function of the human CaV3.3 channel as a result of such interaction. Specifically, we proposed that a combination of polar interactions involving the three hydroxyl groups of genistein and an aromatic interaction with the fused rings are the main binding interactions in the complex formation. Our results pave the way for the rational development of improved and novel low-voltage activated T-type calcium channel inhibitors.
Collapse
Affiliation(s)
- Maricruz Rangel-Galván
- School of Chemical Sciences, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla 72570, Mexico
| | - Azahel Rangel
- Coordinación Académica Región Altiplano, Universidad Autónoma de San Luis Potosí, San Luis Potosí 78700, México
| | - Catalina Romero-Méndez
- Departamento de Fisiología y Biofísica, Facultad de Medicina Universidad Autónoma de San Luis Potosí, San Luis Potosí 78210, México
| | - Eliud Morales Dávila
- School of Chemical Sciences, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla 72570, Mexico
| | - María Eugenia Castro
- Chemistry Center, Science Institute, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla 72570, Mexico
| | - Norma A. Caballero
- School of Biological Sciences, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla 72570, Mexico
| | | | - Brenda L. Sanchez-Gaytan
- Chemistry Center, Science Institute, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla 72570, Mexico
| | - Ulises Meza
- Departamento de Fisiología y Biofísica, Facultad de Medicina Universidad Autónoma de San Luis Potosí, San Luis Potosí 78210, México
| | - Jose Manuel Perez-Aguilar
- School of Chemical Sciences, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla 72570, Mexico
| |
Collapse
|
15
|
Jensterle M, Rizzo M, Janez A. Glucagon-Like Peptide 1 and Taste Perception: From Molecular Mechanisms to Potential Clinical Implications. Int J Mol Sci 2021; 22:ijms22020902. [PMID: 33477478 PMCID: PMC7830704 DOI: 10.3390/ijms22020902] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/03/2021] [Accepted: 01/15/2021] [Indexed: 12/14/2022] Open
Abstract
Preclinical studies provided some important insights into the action of glucagon-like peptide 1 (GLP-1) in taste perception. This review examines the literature to uncover some molecular mechanisms and connections between GLP-1 and the gustatory coding. Local GLP-1 production in the taste bud cells, the expression of GLP-1 receptor on the adjacent nerves, a functional continuum in the perception of sweet chemicals from the gut to the tongue and an identification of GLP-1 induced signaling pathways in peripheral and central gustatory coding all strongly suggest that GLP-1 is involved in the taste perception, especially sweet. However, the impact of GLP-1 based therapies on gustatory coding in humans remains largely unaddressed. Based on the molecular background we encourage further exploration of the tongue as a new treatment target for GLP-1 receptor agonists in clinical studies. Given that pharmacological manipulation of gustatory coding may represent a new potential strategy against obesity and diabetes, the topic is of utmost clinical relevance.
Collapse
Affiliation(s)
- Mojca Jensterle
- Diabetes and Metabolic Diseases, Division of Internal Medicine, Department of Endocrinology, University Medical Centre Ljubljana, Zaloška Cesta 7, 1000 Ljubljana, Slovenia;
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Zaloška Cesta 7, 1000 Ljubljana, Slovenia
| | - Manfredi Rizzo
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of South Carolina, Columbia, SC 29208, USA;
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90133 Palermo, Italy
| | - Andrej Janez
- Diabetes and Metabolic Diseases, Division of Internal Medicine, Department of Endocrinology, University Medical Centre Ljubljana, Zaloška Cesta 7, 1000 Ljubljana, Slovenia;
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, Zaloška Cesta 7, 1000 Ljubljana, Slovenia
- Correspondence: ; Tel.: +386-1-522-3114; Fax: +386-1-522-9359
| |
Collapse
|
16
|
Ahmad R, Dalziel JE. G Protein-Coupled Receptors in Taste Physiology and Pharmacology. Front Pharmacol 2020; 11:587664. [PMID: 33390961 PMCID: PMC7774309 DOI: 10.3389/fphar.2020.587664] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/09/2020] [Indexed: 12/14/2022] Open
Abstract
Heterotrimeric G protein-coupled receptors (GPCRs) comprise the largest receptor family in mammals and are responsible for the regulation of most physiological functions. Besides mediating the sensory modalities of olfaction and vision, GPCRs also transduce signals for three basic taste qualities of sweet, umami (savory taste), and bitter, as well as the flavor sensation kokumi. Taste GPCRs reside in specialised taste receptor cells (TRCs) within taste buds. Type I taste GPCRs (TAS1R) form heterodimeric complexes that function as sweet (TAS1R2/TAS1R3) or umami (TAS1R1/TAS1R3) taste receptors, whereas Type II are monomeric bitter taste receptors or kokumi/calcium-sensing receptors. Sweet, umami and kokumi receptors share structural similarities in containing multiple agonist binding sites with pronounced selectivity while most bitter receptors contain a single binding site that is broadly tuned to a diverse array of bitter ligands in a non-selective manner. Tastant binding to the receptor activates downstream secondary messenger pathways leading to depolarization and increased intracellular calcium in TRCs, that in turn innervate the gustatory cortex in the brain. Despite recent advances in our understanding of the relationship between agonist binding and the conformational changes required for receptor activation, several major challenges and questions remain in taste GPCR biology that are discussed in the present review. In recent years, intensive integrative approaches combining heterologous expression, mutagenesis and homology modeling have together provided insight regarding agonist binding site locations and molecular mechanisms of orthosteric and allosteric modulation. In addition, studies based on transgenic mice, utilizing either global or conditional knock out strategies have provided insights to taste receptor signal transduction mechanisms and their roles in physiology. However, the need for more functional studies in a physiological context is apparent and would be enhanced by a crystallized structure of taste receptors for a more complete picture of their pharmacological mechanisms.
Collapse
Affiliation(s)
- Raise Ahmad
- Food Nutrition and Health Team, Food and Bio-based Products Group, AgResearch, Palmerston North, New Zealand
| | - Julie E Dalziel
- Food Nutrition and Health Team, Food and Bio-based Products Group, AgResearch, Palmerston North, New Zealand
| |
Collapse
|
17
|
Arodola OA, Kanchi S, Hloma P, Bisetty K, Asiri AM, Inamuddin. An in-silico layer-by-layer adsorption study of the interaction between Rebaudioside A and the T1R2 human sweet taste receptor: modelling and biosensing perspectives. Sci Rep 2020; 10:18391. [PMID: 33110140 PMCID: PMC7591876 DOI: 10.1038/s41598-020-75123-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 09/28/2020] [Indexed: 12/03/2022] Open
Abstract
The human sweet taste receptor (T1R2) monomer-a member of the G-protein coupled receptor family that detects a wide variety of chemically and structurally diverse sweet tasting molecules, is known to pose a significant threat to human health. Protein that lack crystal structure is a challenge in structure-based protein design. This study focused on the interaction of the T1R2 monomer with rebaudioside A (Reb-A), a steviol glycoside with potential use as a natural sweetener using in-silico and biosensing methods. Herein, homology modelling, docking studies, and molecular dynamics simulations were applied to elucidate the interaction between Reb-A and the T1R2 monomer. In addition, the electrochemical sensing of the immobilised T1R2-Reb-A complex with zinc oxide nanoparticles (ZnONPs) and graphene oxide (GO) were assessed by testing the performance of multiwalled carbon nanotube (MWCNT) as an adsorbent experimentally. Results indicate a strong interaction between Reb-A and the T1R2 receptor, revealing the stabilizing interaction of the amino acids with the Reb-A by hydrogen bonds with the hydroxyl groups of the glucose moieties, along with a significant amount of hydrophobic interactions. Moreover, the presence of the MWCNT as an anchor confirms the adsorption strength of the T1R2-Reb-A complex onto the GO nanocomposite and supported with electrochemical measurements. Overall, this study could serve as a cornerstone in the development of electrochemical immunosensor for the detection of Reb-A, with applications in the food industry.
Collapse
Affiliation(s)
- Olayide A Arodola
- Department of Chemistry, Durban University of Technology, P.O Box 1334, Durban, 4000, South Africa.
| | - Suvardhan Kanchi
- Department of Chemistry, Durban University of Technology, P.O Box 1334, Durban, 4000, South Africa
| | - Phathisanani Hloma
- Department of Chemistry, Durban University of Technology, P.O Box 1334, Durban, 4000, South Africa
| | - Krishna Bisetty
- Department of Chemistry, Durban University of Technology, P.O Box 1334, Durban, 4000, South Africa.
| | - Abdullah M Asiri
- Chemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Inamuddin
- Chemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia.
- Advanced Functional Materials Laboratory, Department of Applied Chemistry, Faculty of Engineering and Technology, Aligarh Muslim University, Aligarh, 202 002, India.
| |
Collapse
|
18
|
Podlewska S, Bugno R, Kudla L, Bojarski AJ, Przewlocki R. Molecular Modeling of µ Opioid Receptor Ligands with Various Functional Properties: PZM21, SR-17018, Morphine, and Fentanyl-Simulated Interaction Patterns Confronted with Experimental Data. Molecules 2020; 25:E4636. [PMID: 33053718 PMCID: PMC7594085 DOI: 10.3390/molecules25204636] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/04/2020] [Accepted: 10/06/2020] [Indexed: 11/16/2022] Open
Abstract
Molecular modeling approaches are an indispensable part of the drug design process. They not only support the process of searching for new ligands of a given receptor, but they also play an important role in explaining particular activity pathways of a compound. In this study, a comprehensive molecular modeling protocol was developed to explain the observed activity profiles of selected µ opioid receptor agents: two G protein-biased µ opioid receptor agonists(PZM21 and SR-17018), unbiased morphine, and the β-arrestin-2-biased agonist,fentanyl. The study involved docking and molecular dynamics simulations carried out for three crystal structures of the target at a microsecond scale, followed by the statistical analysis of ligand-protein contacts. The interaction frequency between the modeled compounds and the subsequent residues of a protein during the simulation was also correlated with the output of in vitro and in vivo tests, resulting in the set of amino acids with the highest Pearson correlation coefficient values. Such indicated positions may serve as a guide for designing new G protein-biased ligands of the µ opioid receptor.
Collapse
Affiliation(s)
- Sabina Podlewska
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Cracow, Poland;
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Cracow, Poland; (R.B.); (L.K.); (A.J.B.)
| | - Ryszard Bugno
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Cracow, Poland; (R.B.); (L.K.); (A.J.B.)
| | - Lucja Kudla
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Cracow, Poland; (R.B.); (L.K.); (A.J.B.)
| | - Andrzej J. Bojarski
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Cracow, Poland; (R.B.); (L.K.); (A.J.B.)
| | - Ryszard Przewlocki
- Maj Institute of Pharmacology, Polish Academy of Sciences, 12 Smętna Street, 31-343 Cracow, Poland; (R.B.); (L.K.); (A.J.B.)
| |
Collapse
|
19
|
Spaggiari G, Di Pizio A, Cozzini P. Sweet, umami and bitter taste receptors: State of the art of in silico molecular modeling approaches. Trends Food Sci Technol 2020. [DOI: 10.1016/j.tifs.2019.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|