1
|
Zhuang Y, Howard RJ, Lindahl E. Symmetry-adapted Markov state models of closing, opening, and desensitizing in α 7 nicotinic acetylcholine receptors. Nat Commun 2024; 15:9022. [PMID: 39424796 PMCID: PMC11489734 DOI: 10.1038/s41467-024-53170-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 10/03/2024] [Indexed: 10/21/2024] Open
Abstract
α7 nicotinic acetylcholine receptors (nAChRs) are homopentameric ligand-gated ion channels with critical roles in the nervous system. Recent studies have resolved and functionally annotated closed, open, and desensitized states of these receptors, providing insight into ion permeation and lipid binding. However, the process by which α7 nAChRs transition between states remains unclear. To understand gating and lipid modulation, we generated two ensembles of molecular dynamics simulations of apo α7 nAChRs, with or without cholesterol. Using symmetry-adapted Markov state modeling, we developed a five-state gating model. Free energies recapitulated functional behavior, with the closed state dominating in absence of agonist. Open-to-nonconducting transition rates corresponded to experimental open durations. Cholesterol relatively stabilized the desensitized state, and reduced open-desensitized barriers. These results establish plausible asymmetric transition pathways between states, define lipid modulation effects on the α7 nAChR conformational cycle, and provide an ensemble of structural models applicable to rational design of lipidic pharmaceuticals.
Collapse
Affiliation(s)
- Yuxuan Zhuang
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Stockholm, Sweden
| | - Rebecca J Howard
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Stockholm, Sweden
| | - Erik Lindahl
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Stockholm, Sweden.
- Department of Applied Physics, Swedish e-Science Research Center, KTH Royal Institute of Technology, Solna, Stockholm, Sweden.
| |
Collapse
|
2
|
Geoffroy C, Berraud-Pache R, Chéron N, McCort-Tranchepain I, Doria J, Paoletti P, Mony L. Reversible Control of Native GluN2B-Containing NMDA Receptors with Visible Light. ACS Chem Neurosci 2024; 15:3321-3343. [PMID: 39242213 DOI: 10.1021/acschemneuro.4c00247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2024] Open
Abstract
NMDA receptors (NMDARs) are glutamate-gated ion channels playing a central role in synaptic transmission and plasticity. NMDAR dysregulation is linked to various neuropsychiatric disorders. This is particularly true for GluN2B-containing NMDARs (GluN2B-NMDARs), which have major pro-cognitive, but also pro-excitotoxic roles, although their exact involvement in these processes remains debated. Traditional GluN2B-selective antagonists suffer from slow and irreversible effects, limiting their use in native tissues. We therefore developed OptoNAM-3, a photoswitchable negative allosteric modulator selective for GluN2B-NMDARs. OptoNAM-3 provided light-induced reversible inhibition of GluN2B-NMDAR activity with precise temporal control both in vitro and in vivo on the behavior of freely moving Xenopus tadpoles. When bound to GluN2B-NMDARs, OptoNAM-3 displayed remarkable red-shifting of its photoswitching properties allowing the use of blue light instead of UV light to turn-off its activity, which we attributed to geometric constraints imposed by the binding site onto the azobenzene moiety of the ligand. This study therefore highlights the importance of the binding site in shaping the photochemical properties of azobenzene-based photoswitches. In addition, by enabling selective, fast, and reversible photocontrol of native GluN2B-NMDARs with in vivo compatible photochemical properties (visible light), OptoNAM-3 should be a useful tool for the investigation of the GluN2B-NMDAR physiology in native tissues.
Collapse
Affiliation(s)
- Chloé Geoffroy
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Paris F-75005, France
| | - Romain Berraud-Pache
- Laboratoire d'Archéologie Moléculaire et Structurale (LAMS), CNRS UMR 8220, Sorbonne Université, Paris 75005, France
| | - Nicolas Chéron
- PASTEUR, Département de chimie, École normale supérieure, CNRS, Université PSL, Sorbonne Université, Paris 75005, France
| | - Isabelle McCort-Tranchepain
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, CNRS UMR8601, Université Paris Cité, Paris 75006, France
| | - Julia Doria
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Paris F-75005, France
| | - Pierre Paoletti
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Paris F-75005, France
| | - Laetitia Mony
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Paris F-75005, France
| |
Collapse
|
3
|
Qin W, Deng Y, Ren H, Liu Y, Liu L, Liu W, Zhao Y, Li C, Yang Z. Exploring the anticancer mechanism of cardiac glycosides using proteome integral solubility alteration approach. Cancer Med 2024; 13:e70252. [PMID: 39350574 PMCID: PMC11442762 DOI: 10.1002/cam4.70252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/08/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND AND AIMS Cardiac glycosides (CGs), traditionally used for heart failure, have shown potential as anti-cancer agents. This study aims to explore their multifaceted mechanisms in cancer cell biology using proteome integral solubility alteration (PISA), focusing on the interaction with key proteins implicated in cellular metabolism and mitochondrial function. METHODS We conducted lysate-based and intact-cell PISA assays on cancer cells treated with CGs (Digoxin, Digitoxin, Ouabain) to analyze protein solubility changes. This was followed by mass spectrometric analysis and bioinformatics to identify differentially soluble proteins (DSPs). Molecular docking simulations were performed to predict protein-CG interactions. Public data including gene expression changes upon CG treatment were re-analyzed for validation. RESULTS The PISA assays revealed CGs' broad-spectrum interactions, particularly affecting proteins like PKM2, ANXA2, SLC16A1, GOT2 and GLUD1. Molecular docking confirmed stable interactions between CGs and these DSPs. Re-analysis of public data supported the impact of CGs on cancer metabolism and cell signaling pathways. CONCLUSION Our findings suggest that CGs could be repurposed for cancer therapy by modulating cellular processes. The PISA data provide insights into the polypharmacological effects of CGs, warranting further exploration of their mechanisms and clinical potential.
Collapse
Affiliation(s)
- Wenjie Qin
- Department of PharmacyThe First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital)ChangshaChina
| | - Yinhua Deng
- Department of PharmacyThe First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital)ChangshaChina
| | - Huan Ren
- Department of PharmacyThe First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital)ChangshaChina
| | - Yanling Liu
- Department of PharmacyThe First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital)ChangshaChina
| | - Ling Liu
- Department of PharmacyThe First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital)ChangshaChina
| | - Wenhui Liu
- Department of PharmacyThe Second Xiangya Hospital, Central South UniversityChangshaChina
- Institute of Clinical Pharmacy, Central South UniversityChangshaChina
| | - Yuxi Zhao
- Shenzhen Wininnovate Bio‐Tech Co., LtdShenzhenChina
| | - Chen Li
- Department of PharmacyThe First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital)ChangshaChina
| | - Zhiling Yang
- Department of PharmacyThe First Affiliated Hospital of Hunan Normal University (Hunan Provincial People's Hospital)ChangshaChina
| |
Collapse
|
4
|
Habibpour M, Razaghi-Moghadam Z, Nikoloski Z. Machine learning of metabolite-protein interactions from model-derived metabolic phenotypes. NAR Genom Bioinform 2024; 6:lqae114. [PMID: 39229256 PMCID: PMC11369697 DOI: 10.1093/nargab/lqae114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/10/2024] [Accepted: 08/29/2024] [Indexed: 09/05/2024] Open
Abstract
Unraveling metabolite-protein interactions is key to identifying the mechanisms by which metabolism affects the function of other cellular layers. Despite extensive experimental and computational efforts to identify the regulatory roles of metabolites in interaction with proteins, it remains challenging to achieve a genome-scale coverage of these interactions. Here, we leverage established gold standards for metabolite-protein interactions to train supervised classifiers using features derived from genome-scale metabolic models and matched data on protein abundance and reaction fluxes to distinguish interacting from non-interacting pairs. Through a comprehensive comparative study, we explore the impact of different features and assess the effect of gold standards for non-interacting pairs on the performance of the classifiers. Using data sets from Escherichia coli and Saccharomyces cerevisiae, we demonstrate that the features constructed by integrating fluxomic and proteomic data with metabolic phenotypes predicted from genome-scale metabolic models can be effectively used to train classifiers, accurately predicting metabolite-protein interactions in the context of metabolism. Our results reveal that the high performance of classifiers trained on these features is unaffected by the method used to generate gold standards for non-interacting pairs. Overall, our study introduces valuable features that improve the performance of identifying metabolite-protein interactions in the context of metabolism.
Collapse
Affiliation(s)
- Mahdis Habibpour
- Bioinformatics Department, Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Zahra Razaghi-Moghadam
- Bioinformatics Department, Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
- Systems Biology and Mathematical Modeling, Max Planck Institute of Molecular Plant Physiology, Potsdam, Germany
| | - Zoran Nikoloski
- Bioinformatics Department, Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
- Systems Biology and Mathematical Modeling, Max Planck Institute of Molecular Plant Physiology, Potsdam, Germany
| |
Collapse
|
5
|
Brunori M. Wandering about allostery. Biol Direct 2024; 19:64. [PMID: 39113091 PMCID: PMC11308221 DOI: 10.1186/s13062-024-00502-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 08/10/2024] Open
Abstract
It was a Lucky Strike to be working with Eraldo Antonini on hemoglobin and myoglobin when Jeffries Wyman arrived in Rome in 1961. I found myself connected with a number of creative scientists when the concept of allosteric control was conceived and gifted to the life science community. In retrospect, this was a demonstration of the skill and imagination of a few intelligent scientists that I happened to be close to. Those talents demonstrated the power of creativity as pictured by the motto "Mens agitat molem"; a celebration of humanism and intellect that paved the way to novel discoveries in the field of structure function relationships in proteins. I have presented hereby some of the events and the people as emerged from my memory over three decades of exciting scientific life.
Collapse
Affiliation(s)
- Maurizio Brunori
- Dipartimento di Scienze Biochimiche, Presidente emerito della Classe di Scienze FMN, Accademia Nazionale dei Lincei; Professore emerito di Chimica e Biochimica, Sapienza Università di Roma, Rome, Italy.
| |
Collapse
|
6
|
Gao X, Yeh HI, Yang Z, Fan C, Jiang F, Howard RJ, Lindahl E, Kappes JC, Hwang TC. Allosteric inhibition of CFTR gating by CFTRinh-172 binding in the pore. Nat Commun 2024; 15:6668. [PMID: 39107303 PMCID: PMC11303713 DOI: 10.1038/s41467-024-50641-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
Loss-of-function mutations of the CFTR gene cause the life-shortening genetic disease cystic fibrosis (CF), whereas overactivity of CFTR may lead to secretory diarrhea and polycystic kidney disease. While effective drugs targeting the CFTR protein have been developed for the treatment of CF, little progress has been made for diseases caused by hyper-activated CFTR. Here, we solve the cryo-EM structure of CFTR in complex with CFTRinh-172 (Inh-172), a CFTR gating inhibitor with promising potency and efficacy. We find that Inh-172 binds inside the pore of CFTR, interacting with amino acid residues from transmembrane segments (TMs) 1, 6, 8, 9, and 12 through mostly hydrophobic interactions and a salt bridge. Substitution of these residues lowers the apparent affinity of Inh-172. The inhibitor-bound structure reveals re-orientations of the extracellular segment of TMs 1, 8, and 12, supporting an allosteric modulation mechanism involving post-binding conformational changes. This allosteric inhibitory mechanism readily explains our observations that pig CFTR, which preserves all the amino acid residues involved in Inh-172 binding, exhibits a much-reduced sensitivity to Inh-172 and that the apparent affinity of Inh-172 is altered by the CF drug ivacaftor (i.e., VX-770) which enhances CFTR's activity through binding to a site also comprising TM8.
Collapse
Affiliation(s)
- Xiaolong Gao
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO, 65211, USA.
| | - Han-I Yeh
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO, 65211, USA
- Institute of Pharmacology, National Yang Ming Chiao Tung University, College of Medicine, Taipei, Taiwan
- Membrane Protein Structural Biology Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Zhengrong Yang
- Heersink School of Medicine, University of Alabama School of Medicine, Birmingham, AL, 35233, USA
| | - Chen Fan
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Fan Jiang
- Heersink School of Medicine, University of Alabama School of Medicine, Birmingham, AL, 35233, USA
| | - Rebecca J Howard
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Erik Lindahl
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Solna, Sweden
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - John C Kappes
- Heersink School of Medicine, University of Alabama School of Medicine, Birmingham, AL, 35233, USA
- Research Service, Birmingham Veterans Affairs Medical Center, Veterans Health Administration, Birmingham, AL, 35233, USA
| | - Tzyh-Chang Hwang
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO, 65211, USA.
- Institute of Pharmacology, National Yang Ming Chiao Tung University, College of Medicine, Taipei, Taiwan.
- Membrane Protein Structural Biology Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
7
|
Cecchini M, Corringer PJ, Changeux JP. The Nicotinic Acetylcholine Receptor and Its Pentameric Homologs: Toward an Allosteric Mechanism of Signal Transduction at the Atomic Level. Annu Rev Biochem 2024; 93:339-366. [PMID: 38346274 DOI: 10.1146/annurev-biochem-030122-033116] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
The nicotinic acetylcholine receptor has served, since its biochemical identification in the 1970s, as a model of an allosteric ligand-gated ion channel mediating signal transition at the synapse. In recent years, the application of X-ray crystallography and high-resolution cryo-electron microscopy, together with molecular dynamic simulations of nicotinic receptors and homologs, have opened a new era in the understanding of channel gating by the neurotransmitter. They reveal, at atomic resolution, the diversity and flexibility of the multiple ligand-binding sites, including recently discovered allosteric modulatory sites distinct from the neurotransmitter orthosteric site, and the conformational dynamics of the activation process as a molecular switch linking these multiple sites. The model emerging from these studies paves the way for a new pharmacology based, first, upon the occurrence of an original mode of indirect allosteric modulation, distinct from a steric competition for a single and rigid binding site, and second, the design of drugs that specifically interact with privileged conformations of the receptor such as agonists, antagonists, and desensitizers. Research on nicotinic receptors is still at the forefront of understanding the mode of action of drugs on the nervous system.
Collapse
Affiliation(s)
- Marco Cecchini
- Institut de Chimie de Strasbourg, CNRS UMR 7177, Université de Strasbourg, Strasbourg, France
| | - Pierre-Jean Corringer
- Channel Receptors Unit, Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Paris, France
| | - Jean-Pierre Changeux
- Department of Neuroscience, Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Paris, France;
| |
Collapse
|
8
|
Liu B, Zhou H, Tan L, Siu KTH, Guan XY. Exploring treatment options in cancer: Tumor treatment strategies. Signal Transduct Target Ther 2024; 9:175. [PMID: 39013849 PMCID: PMC11252281 DOI: 10.1038/s41392-024-01856-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 07/18/2024] Open
Abstract
Traditional therapeutic approaches such as chemotherapy and radiation therapy have burdened cancer patients with onerous physical and psychological challenges. Encouragingly, the landscape of tumor treatment has undergone a comprehensive and remarkable transformation. Emerging as fervently pursued modalities are small molecule targeted agents, antibody-drug conjugates (ADCs), cell-based therapies, and gene therapy. These cutting-edge treatment modalities not only afford personalized and precise tumor targeting, but also provide patients with enhanced therapeutic comfort and the potential to impede disease progression. Nonetheless, it is acknowledged that these therapeutic strategies still harbour untapped potential for further advancement. Gaining a comprehensive understanding of the merits and limitations of these treatment modalities holds the promise of offering novel perspectives for clinical practice and foundational research endeavours. In this review, we discussed the different treatment modalities, including small molecule targeted drugs, peptide drugs, antibody drugs, cell therapy, and gene therapy. It will provide a detailed explanation of each method, addressing their status of development, clinical challenges, and potential solutions. The aim is to assist clinicians and researchers in gaining a deeper understanding of these diverse treatment options, enabling them to carry out effective treatment and advance their research more efficiently.
Collapse
Affiliation(s)
- Beilei Liu
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| | - Hongyu Zhou
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Licheng Tan
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Kin To Hugo Siu
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, China.
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China.
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou, China.
- MOE Key Laboratory of Tumor Molecular Biology, Jinan University, Guangzhou, China.
| |
Collapse
|
9
|
Borghese CM, Goldschen-Ohm MP. State-dependent energetics of GABA A receptor modulators. Biophys J 2024; 123:1903-1906. [PMID: 38303510 PMCID: PMC11309981 DOI: 10.1016/j.bpj.2024.01.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/23/2024] [Accepted: 01/29/2024] [Indexed: 02/03/2024] Open
Affiliation(s)
- Cecilia M Borghese
- University of Texas at Austin, Department of Neuroscience, Austin, Texas
| | | |
Collapse
|
10
|
Lopez-Balastegui M, Stepniewski TM, Kogut-Günthel MM, Di Pizio A, Rosenkilde MM, Mao J, Selent J. Relevance of G protein-coupled receptor (GPCR) dynamics for receptor activation, signalling bias and allosteric modulation. Br J Pharmacol 2024. [PMID: 38978399 DOI: 10.1111/bph.16495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 04/22/2024] [Accepted: 05/23/2024] [Indexed: 07/10/2024] Open
Abstract
G protein-coupled receptors (GPCRs) are one of the major drug targets. In recent years, computational drug design for GPCRs has mainly focused on static structures obtained through X-ray crystallography, cryogenic electron microscopy (cryo-EM) or in silico modelling as a starting point for virtual screening campaigns. However, GPCRs are highly flexible entities with the ability to adopt different conformational states that elicit different physiological responses. Including this knowledge in the drug discovery pipeline can help to tailor novel conformation-specific drugs with an improved therapeutic profile. In this review, we outline our current knowledge about GPCR dynamics that is relevant for receptor activation, signalling bias and allosteric modulation. Ultimately, we highlight new technological implementations such as time-resolved X-ray crystallography and cryo-EM as well as computational algorithms that can contribute to a more comprehensive understanding of receptor dynamics and its relevance for GPCR functionality.
Collapse
Affiliation(s)
- Marta Lopez-Balastegui
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute & Pompeu Fabra University, Barcelona, Spain
| | - Tomasz Maciej Stepniewski
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute & Pompeu Fabra University, Barcelona, Spain
- InterAx Biotech AG, Villigen, Switzerland
| | | | - Antonella Di Pizio
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- Chair for Chemoinformatics and Protein Modelling, Department of Molecular Life Science, School of Science, Technical University of Munich, Freising, Germany
| | - Mette Marie Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences University of Copenhagen, København N, Denmark
| | - Jiafei Mao
- Huairou Research Center, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Jana Selent
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute & Pompeu Fabra University, Barcelona, Spain
| |
Collapse
|
11
|
Wang YJ, Di XJ, Zhang PP, Chen X, Williams MP, Han DY, Nashmi R, Henderson BJ, Moss FJ, Mu TW. Hsp47 promotes biogenesis of multi-subunit neuroreceptors in the endoplasmic reticulum. eLife 2024; 13:e84798. [PMID: 38963323 PMCID: PMC11257679 DOI: 10.7554/elife.84798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 05/21/2024] [Indexed: 07/05/2024] Open
Abstract
Protein homeostasis (proteostasis) deficiency is an important contributing factor to neurological and metabolic diseases. However, how the proteostasis network orchestrates the folding and assembly of multi-subunit membrane proteins is poorly understood. Previous proteomics studies identified Hsp47 (Gene: SERPINH1), a heat shock protein in the endoplasmic reticulum lumen, as the most enriched interacting chaperone for gamma-aminobutyric acid type A (GABAA) receptors. Here, we show that Hsp47 enhances the functional surface expression of GABAA receptors in rat neurons and human HEK293T cells. Furthermore, molecular mechanism study demonstrates that Hsp47 acts after BiP (Gene: HSPA5) and preferentially binds the folded conformation of GABAA receptors without inducing the unfolded protein response in HEK293T cells. Therefore, Hsp47 promotes the subunit-subunit interaction, the receptor assembly process, and the anterograde trafficking of GABAA receptors. Overexpressing Hsp47 is sufficient to correct the surface expression and function of epilepsy-associated GABAA receptor variants in HEK293T cells. Hsp47 also promotes the surface trafficking of other Cys-loop receptors, including nicotinic acetylcholine receptors and serotonin type 3 receptors in HEK293T cells. Therefore, in addition to its known function as a collagen chaperone, this work establishes that Hsp47 plays a critical and general role in the maturation of multi-subunit Cys-loop neuroreceptors.
Collapse
Affiliation(s)
- Ya-Juan Wang
- Department of Physiology and Biophysics, Case Western Reserve UniversityClevelandUnited States
| | - Xiao-Jing Di
- Department of Physiology and Biophysics, Case Western Reserve UniversityClevelandUnited States
| | - Pei-Pei Zhang
- Department of Physiology and Biophysics, Case Western Reserve UniversityClevelandUnited States
| | - Xi Chen
- Department of Physiology and Biophysics, Case Western Reserve UniversityClevelandUnited States
| | - Marnie P Williams
- Department of Physiology and Biophysics, Case Western Reserve UniversityClevelandUnited States
| | - Dong-Yun Han
- Department of Physiology and Biophysics, Case Western Reserve UniversityClevelandUnited States
| | - Raad Nashmi
- Department of Biology, University of VictoriaVictoriaCanada
| | - Brandon J Henderson
- Department of Biomedical Sciences, Marshall UniversityHuntingtonUnited States
| | - Fraser J Moss
- Department of Physiology and Biophysics, Case Western Reserve UniversityClevelandUnited States
| | - Ting-Wei Mu
- Department of Physiology and Biophysics, Case Western Reserve UniversityClevelandUnited States
| |
Collapse
|
12
|
Saito A, Alvi S, Valant C, Christopoulos A, Carbone SE, Poole DP. Therapeutic potential of allosteric modulators for the treatment of gastrointestinal motility disorders. Br J Pharmacol 2024; 181:2232-2246. [PMID: 36565295 DOI: 10.1111/bph.16023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/24/2022] [Accepted: 12/09/2022] [Indexed: 12/25/2022] Open
Abstract
Gastrointestinal motility is tightly regulated by the enteric nervous system (ENS). Disruption of coordinated enteric nervous system activity can result in dysmotility. Pharmacological treatment options for dysmotility include targeting of G protein-coupled receptors (GPCRs) expressed by neurons of the enteric nervous system. Current GPCR-targeting drugs for motility disorders bind to the highly conserved endogenous ligand-binding site and promote indiscriminate activation or inhibition of the target receptor throughout the body. This can be associated with significant side-effect liability and a loss of physiological tone. Allosteric modulators of GPCRs bind to a distinct site from the endogenous ligand, which is typically less conserved across multiple receptor subtypes and can modulate endogenous ligand signalling. Allosteric modulation of GPCRs that are important for enteric nervous system function may provide effective relief from motility disorders while limiting side-effects. This review will focus on how allosteric modulators of GPCRs may influence gastrointestinal motility, using 5-hydroxytryptamine (5-HT), acetylcholine (ACh) and opioid receptors as examples. LINKED ARTICLES: This article is part of a themed issue Therapeutic Targeting of G Protein-Coupled Receptors: hot topics from the Australasian Society of Clinical and Experimental Pharmacologists and Toxicologists 2021 Virtual Annual Scientific Meeting. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.14/issuetoc.
Collapse
Affiliation(s)
- Ayame Saito
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Sadia Alvi
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Celine Valant
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Simona E Carbone
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Daniel P Poole
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| |
Collapse
|
13
|
Healy LD, Fernández JA, Aiolfi R, Mosnier LO, Griffin JH. An orthosteric/allosteric bivalent peptide agonist comprising covalently linked protease-activated receptor-derived peptides mimics in vitro and in vivo activities of activated protein C. J Thromb Haemost 2024; 22:2039-2051. [PMID: 38670314 DOI: 10.1016/j.jtha.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/22/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND Activated protein C (APC) has anticoagulant and cytoprotective cell-signaling activities, which often require protease-activated receptor (PAR) 1 and PAR3 and PAR cleavages at noncanonical sites (R46-N47 and R41-G42, respectively). Some PAR1-derived (P1) peptides and PAR3-derived (P3) peptides, eg, P1-47-66 and P3-42-65, mimic APC's cell signaling. In anti-inflammatory assays, these 2 peptides at low concentrations synergistically attenuate cellular inflammation. OBJECTIVES To determine whether a P1 peptide covalently linked to a P3 peptide mimics APC's anti-inflammatory and endothelial barrier stabilization activities. METHODS Anti-inflammatory assays employed stimulated THP-1 cells and caspase-1 measurements. Cultured human EA.hy926 or murine aortic endothelial cells (ECs) exposed to thrombin were monitored for transendothelial electrical resistance. Bivalent covalently linked P1:P3 peptides were studied for APC-like activities. RESULTS In anti-inflammatory assays, P1-47-55 was as active as P1-47-66 and some P3 peptides (eg, P3-44-54 and P3-51-65) were as active as P3-42-65. The bivalent P1:P3 peptide comprising P1-47-55-(Gly[10 residues])-P3-51-65 (designated "G10 peptide") was more potently anti-inflammatory than the P1 or P3 peptide alone. In transendothelial electrical resistance studies of thrombin-challenged ECs, P1-47-55 and the G10 peptide mimicked APC's protective actions. In dose-response studies, the G10 peptide was more potent than the P1-47-55 peptide. In murine EC studies, the murine PAR-sequence-derived G10 peptide mimicked murine APC's activity. Anti-PAR1 and anti-PAR3 antibodies, but not anti-endothelial protein C receptor antibodies, abated G10's cytoprotection, showing that G10's actions involve PAR1:PAR3. G10 significantly increased survival in murine endotoxemia. CONCLUSION The PAR-sequence-derived G10 peptide is a bivalent agonist that mimics APC's cytoprotective, anti-inflammatory, and endothelial barrier-stabilizing actions and APC's protection against endotoxemic mortality.
Collapse
Affiliation(s)
- Laura D Healy
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - José A Fernández
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Roberto Aiolfi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Laurent O Mosnier
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - John H Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA.
| |
Collapse
|
14
|
Abaffy T, Fu O, Harume-Nagai M, Goldenberg JM, Kenyon V, Kenakin T. Intracellular Allosteric Antagonist of the Olfactory Receptor OR51E2. Mol Pharmacol 2024; 106:21-32. [PMID: 38719475 PMCID: PMC11187688 DOI: 10.1124/molpharm.123.000843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/16/2024] [Indexed: 06/20/2024] Open
Abstract
Olfactory receptors are members of class A (rhodopsin-like) family of G protein-coupled receptors (GPCRs). Their expression and function have been increasingly studied in nonolfactory tissues, and many have been identified as potential therapeutic targets. In this manuscript, we focus on the discovery of novel ligands for the olfactory receptor family 51 subfamily E2 (OR51E2). We performed an artificial intelligence-based virtual drug screen of a ∼2.2 million small molecule library. Cell-based functional assay identified compound 80 (C80) as an antagonist and inverse agonist, and detailed pharmacological analysis revealed C80 acts as a negative allosteric modulator by significantly decreasing the agonist efficacy, while having a minimal effect on receptor affinity for agonist. C80 binds to an allosteric binding site formed by a network of nine residues localized in the intracellular parts of transmembrane domains 3, 5, 6, 7, and H8, which also partially overlaps with a G protein binding site. Mutational experiments of residues involved in C80 binding uncovered the significance of the C2406.37 position in blocking the activation-related conformational change and keeping the receptor in the inactive form. Our study provides a mechanistic understanding of the negative allosteric action of C80 on agonist-ctivated OR51E2. We believe the identification of the antagonist of OR51E2 will enable a multitude of studies aiming to determine the functional role of this receptor in specific biologic processes. SIGNIFICANCE STATEMENT: OR51E2 has been implicated in various biological processes, and its antagonists that can effectively modulate its activity have therapeutic potential. Here we report the discovery of a negative allosteric modulator of OR51E2 and provide a mechanistic understanding of its action. We demonstrate that this modulator has an inhibitory effect on the efficacy of the agonist for the receptor and reveal a network of nine residues that constitute its binding pocket, which also partially overlaps with the G protein binding site.
Collapse
Affiliation(s)
- Tatjana Abaffy
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, North Carolina (T.A., O.F.); Columbia Center for Human Development/Columbia Center for Stem Cell Therapies Department, Columbia University, New York (M.H.-N.); Chemistry Department, School of Math and Science at the United States Naval Academy, Annapolis, Maryland (J.M.G.); Atomwise Inc., San Francisco, California (J.M.G., V.K.); and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (T.K.)
| | - Olivia Fu
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, North Carolina (T.A., O.F.); Columbia Center for Human Development/Columbia Center for Stem Cell Therapies Department, Columbia University, New York (M.H.-N.); Chemistry Department, School of Math and Science at the United States Naval Academy, Annapolis, Maryland (J.M.G.); Atomwise Inc., San Francisco, California (J.M.G., V.K.); and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (T.K.)
| | - Maira Harume-Nagai
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, North Carolina (T.A., O.F.); Columbia Center for Human Development/Columbia Center for Stem Cell Therapies Department, Columbia University, New York (M.H.-N.); Chemistry Department, School of Math and Science at the United States Naval Academy, Annapolis, Maryland (J.M.G.); Atomwise Inc., San Francisco, California (J.M.G., V.K.); and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (T.K.)
| | - Josh M Goldenberg
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, North Carolina (T.A., O.F.); Columbia Center for Human Development/Columbia Center for Stem Cell Therapies Department, Columbia University, New York (M.H.-N.); Chemistry Department, School of Math and Science at the United States Naval Academy, Annapolis, Maryland (J.M.G.); Atomwise Inc., San Francisco, California (J.M.G., V.K.); and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (T.K.)
| | - Victor Kenyon
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, North Carolina (T.A., O.F.); Columbia Center for Human Development/Columbia Center for Stem Cell Therapies Department, Columbia University, New York (M.H.-N.); Chemistry Department, School of Math and Science at the United States Naval Academy, Annapolis, Maryland (J.M.G.); Atomwise Inc., San Francisco, California (J.M.G., V.K.); and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (T.K.)
| | - Terry Kenakin
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, North Carolina (T.A., O.F.); Columbia Center for Human Development/Columbia Center for Stem Cell Therapies Department, Columbia University, New York (M.H.-N.); Chemistry Department, School of Math and Science at the United States Naval Academy, Annapolis, Maryland (J.M.G.); Atomwise Inc., San Francisco, California (J.M.G., V.K.); and Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (T.K.)
| |
Collapse
|
15
|
Martinez-Corral R, Nam KM, DePace AH, Gunawardena J. The Hill function is the universal Hopfield barrier for sharpness of input-output responses. Proc Natl Acad Sci U S A 2024; 121:e2318329121. [PMID: 38787881 PMCID: PMC11145184 DOI: 10.1073/pnas.2318329121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
The Hill functions, [Formula: see text], have been widely used in biology for over a century but, with the exception of [Formula: see text], they have had no justification other than as a convenient fit to empirical data. Here, we show that they are the universal limit for the sharpness of any input-output response arising from a Markov process model at thermodynamic equilibrium. Models may represent arbitrary molecular complexity, with multiple ligands, internal states, conformations, coregulators, etc, under core assumptions that are detailed in the paper. The model output may be any linear combination of steady-state probabilities, with components other than the chosen input ligand held constant. This formulation generalizes most of the responses in the literature. We use a coarse-graining method in the graph-theoretic linear framework to show that two sharpness measures for input-output responses fall within an effectively bounded region of the positive quadrant, [Formula: see text], for any equilibrium model with [Formula: see text] input binding sites. [Formula: see text] exhibits a cusp which approaches, but never exceeds, the sharpness of [Formula: see text], but the region and the cusp can be exceeded when models are taken away from thermodynamic equilibrium. Such fundamental thermodynamic limits are called Hopfield barriers, and our results provide a biophysical justification for the Hill functions as the universal Hopfield barriers for sharpness. Our results also introduce an object, [Formula: see text], whose structure may be of mathematical interest, and suggest the importance of characterizing Hopfield barriers for other forms of cellular information processing.
Collapse
Affiliation(s)
| | - Kee-Myoung Nam
- Department of Systems Biology, Harvard Medical School, Boston, MA02115
| | - Angela H. DePace
- Department of Systems Biology, Harvard Medical School, Boston, MA02115
- HHMI, Boston, MA02115
| | | |
Collapse
|
16
|
Brunetti L, Francavilla F, Leopoldo M, Lacivita E. Allosteric Modulators of Serotonin Receptors: A Medicinal Chemistry Survey. Pharmaceuticals (Basel) 2024; 17:695. [PMID: 38931362 PMCID: PMC11206742 DOI: 10.3390/ph17060695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) is a neurotransmitter regulating numerous physiological functions, and its dysregulation is a crucial component of the pathological processes of schizophrenia, depression, migraines, and obesity. 5-HT interacts with 14 different receptors, of which 5-HT1A-1FRs, 5-HT2A-CRs, and 5-HT4-7Rs are G protein-coupled receptors (GPCRs), while 5-HT3R is a ligand-gated ion channel. Over the years, selective orthosteric ligands have been identified for almost all serotonin receptors, yielding several clinically relevant drugs. However, the high degree of homology between 5-HTRs and other GPCRs means that orthosteric ligands can have severe side effects. Thus, there has recently been increased interest in developing safer ligands of GPCRs, which bind to less conserved, more specific sites, distinct from that of the receptor's natural ligand. The present review describes the identification of allosteric ligands of serotonin receptors, which are largely natural compounds (oleamide, cannabidiol, THC, and aporphine alkaloids), complemented by synthetic modulators developed in large part for the 5-HT2C receptor. The latter are positive allosteric modulators sought after for their potential as drugs preferable over the orthosteric agonists as antiobesity agents for their potentially safer profile. When available, details on the interactions between the ligand and allosteric binding site will be provided. An outlook on future research in the field will also be provided.
Collapse
Affiliation(s)
| | | | - Marcello Leopoldo
- Department of Pharmacy–Drug Sciences, University of Bari Aldo Moro, 70125 Bari, Italy; (L.B.); (F.F.); (E.L.)
| | | |
Collapse
|
17
|
Gardner EC, Tramont C, Bachanová P, Wang C, Do H, Boutz DR, Kar S, Zemelman BV, Gollihar JD, Ellington AD. Engineering a human P2X2 receptor with altered ligand selectivity in yeast. J Biol Chem 2024; 300:107248. [PMID: 38556082 PMCID: PMC11063903 DOI: 10.1016/j.jbc.2024.107248] [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: 05/29/2023] [Revised: 03/14/2024] [Accepted: 03/23/2024] [Indexed: 04/02/2024] Open
Abstract
P2X receptors are a family of ligand gated ion channels found in a range of eukaryotic species including humans but are not naturally present in the yeast Saccharomyces cerevisiae. We demonstrate the first recombinant expression and functional gating of the P2X2 receptor in baker's yeast. We leverage the yeast host for facile genetic screens of mutant P2X2 by performing site saturation mutagenesis at residues of interest, including SNPs implicated in deafness and at residues involved in native binding. Deep mutational analysis and rounds of genetic engineering yield mutant P2X2 F303Y A304W, which has altered ligand selectivity toward the ATP analog AMP-PNP. The F303Y A304W variant shows over 100-fold increased intracellular calcium amplitudes with AMP-PNP compared to the WT receptor and has a much lower desensitization rate. Since AMP-PNP does not naturally activate P2X receptors, the F303Y A304W P2X2 may be a starting point for downstream applications in chemogenetic cellular control. Interestingly, the A304W mutation selectively destabilizes the desensitized state, which may provide a mechanistic basis for receptor opening with suboptimal agonists. The yeast system represents an inexpensive, scalable platform for ion channel characterization and engineering by circumventing the more expensive and time-consuming methodologies involving mammalian hosts.
Collapse
Affiliation(s)
- Elizabeth C Gardner
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Caitlin Tramont
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Petra Bachanová
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Chad Wang
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Hannah Do
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Daniel R Boutz
- Antibody Discovery and Accelerated Protein Therapeutics, Department of Pathology & Genomic Medicine, Houston Methodist Research Institute, Houston, Texas, USA
| | - Shaunak Kar
- Antibody Discovery and Accelerated Protein Therapeutics, Department of Pathology & Genomic Medicine, Houston Methodist Research Institute, Houston, Texas, USA
| | - Boris V Zemelman
- Department of Neuroscience, Center for Learning and Memory, The University of Texas at Austin, Austin, Texas, USA.
| | - Jimmy D Gollihar
- Antibody Discovery and Accelerated Protein Therapeutics, Department of Pathology & Genomic Medicine, Houston Methodist Research Institute, Houston, Texas, USA.
| | - Andrew D Ellington
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, Texas, USA.
| |
Collapse
|
18
|
Flavin A, Azizi P, Murataeva N, Yust K, Du W, Ross R, Greig I, Nguyen T, Zhang Y, Mackie K, Straiker A. CB1 Receptor Negative Allosteric Modulators as a Potential Tool to Reverse Cannabinoid Toxicity. Molecules 2024; 29:1881. [PMID: 38675703 PMCID: PMC11053441 DOI: 10.3390/molecules29081881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
While the opioid crisis has justifiably occupied news headlines, emergency rooms are seeing many thousands of visits for another cause: cannabinoid toxicity. This is partly due to the spread of cheap and extremely potent synthetic cannabinoids that can cause serious neurological and cardiovascular complications-and deaths-every year. While an opioid overdose can be reversed by naloxone, there is no analogous treatment for cannabis toxicity. Without an antidote, doctors rely on sedatives, with their own risks, or 'waiting it out' to treat these patients. We have shown that the canonical synthetic 'designer' cannabinoids are highly potent CB1 receptor agonists and, as a result, competitive antagonists may struggle to rapidly reverse an overdose due to synthetic cannabinoids. Negative allosteric modulators (NAMs) have the potential to attenuate the effects of synthetic cannabinoids without having to directly compete for binding. We tested a group of CB1 NAMs for their ability to reverse the effects of the canonical synthetic designer cannabinoid JWH018 in vitro in a neuronal model of endogenous cannabinoid signaling and also in vivo. We tested ABD1085, RTICBM189, and PSNCBAM1 in autaptic hippocampal neurons that endogenously express a retrograde CB1-dependent circuit that inhibits neurotransmission. We found that all of these compounds blocked/reversed JWH018, though some proved more potent than others. We then tested whether these compounds could block the effects of JWH018 in vivo, using a test of nociception in mice. We found that only two of these compounds-RTICBM189 and PSNCBAM1-blocked JWH018 when applied in advance. The in vitro potency of a compound did not predict its in vivo potency. PSNCBAM1 proved to be the more potent of the compounds and also reversed the effects of JWH018 when applied afterward, a condition that more closely mimics an overdose situation. Lastly, we found that PSNCBAM1 did not elicit withdrawal after chronic JWH018 treatment. In summary, CB1 NAMs can, in principle, reverse the effects of the canonical synthetic designer cannabinoid JWH018 both in vitro and in vivo, without inducing withdrawal. These findings suggest a novel pharmacological approach to at last provide a tool to counter cannabinoid toxicity.
Collapse
Affiliation(s)
- Audrey Flavin
- Gill Center for Biomolecular Science, Program in Neuroscience, Department of Psychological and Brain Sciences Indiana University, Bloomington, IN 47405, USA (N.M.); (K.Y.); (W.D.); (K.M.)
| | - Paniz Azizi
- Gill Center for Biomolecular Science, Program in Neuroscience, Department of Psychological and Brain Sciences Indiana University, Bloomington, IN 47405, USA (N.M.); (K.Y.); (W.D.); (K.M.)
| | - Natalia Murataeva
- Gill Center for Biomolecular Science, Program in Neuroscience, Department of Psychological and Brain Sciences Indiana University, Bloomington, IN 47405, USA (N.M.); (K.Y.); (W.D.); (K.M.)
| | - Kyle Yust
- Gill Center for Biomolecular Science, Program in Neuroscience, Department of Psychological and Brain Sciences Indiana University, Bloomington, IN 47405, USA (N.M.); (K.Y.); (W.D.); (K.M.)
| | - Wenwen Du
- Gill Center for Biomolecular Science, Program in Neuroscience, Department of Psychological and Brain Sciences Indiana University, Bloomington, IN 47405, USA (N.M.); (K.Y.); (W.D.); (K.M.)
| | - Ruth Ross
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5G 2C8, Canada;
| | - Iain Greig
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK;
| | - Thuy Nguyen
- Research Triangle Institute, Durham, NC 27709, USA; (T.N.); (Y.Z.)
| | - Yanan Zhang
- Research Triangle Institute, Durham, NC 27709, USA; (T.N.); (Y.Z.)
| | - Ken Mackie
- Gill Center for Biomolecular Science, Program in Neuroscience, Department of Psychological and Brain Sciences Indiana University, Bloomington, IN 47405, USA (N.M.); (K.Y.); (W.D.); (K.M.)
| | - Alex Straiker
- Gill Center for Biomolecular Science, Program in Neuroscience, Department of Psychological and Brain Sciences Indiana University, Bloomington, IN 47405, USA (N.M.); (K.Y.); (W.D.); (K.M.)
| |
Collapse
|
19
|
Zhang M, Chen T, Lu X, Lan X, Chen Z, Lu S. G protein-coupled receptors (GPCRs): advances in structures, mechanisms, and drug discovery. Signal Transduct Target Ther 2024; 9:88. [PMID: 38594257 PMCID: PMC11004190 DOI: 10.1038/s41392-024-01803-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/19/2024] [Accepted: 03/13/2024] [Indexed: 04/11/2024] Open
Abstract
G protein-coupled receptors (GPCRs), the largest family of human membrane proteins and an important class of drug targets, play a role in maintaining numerous physiological processes. Agonist or antagonist, orthosteric effects or allosteric effects, and biased signaling or balanced signaling, characterize the complexity of GPCR dynamic features. In this study, we first review the structural advancements, activation mechanisms, and functional diversity of GPCRs. We then focus on GPCR drug discovery by revealing the detailed drug-target interactions and the underlying mechanisms of orthosteric drugs approved by the US Food and Drug Administration in the past five years. Particularly, an up-to-date analysis is performed on available GPCR structures complexed with synthetic small-molecule allosteric modulators to elucidate key receptor-ligand interactions and allosteric mechanisms. Finally, we highlight how the widespread GPCR-druggable allosteric sites can guide structure- or mechanism-based drug design and propose prospects of designing bitopic ligands for the future therapeutic potential of targeting this receptor family.
Collapse
Affiliation(s)
- Mingyang Zhang
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ting Chen
- Department of Cardiology, Changzheng Hospital, Affiliated to Naval Medical University, Shanghai, 200003, China
| | - Xun Lu
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaobing Lan
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China
| | - Ziqiang Chen
- Department of Orthopedics, Changhai Hospital, Affiliated to Naval Medical University, Shanghai, 200433, China.
| | - Shaoyong Lu
- Key Laboratory of Protection, Development and Utilization of Medicinal Resources in Liupanshan Area, Ministry of Education, Peptide & Protein Drug Research Center, School of Pharmacy, Ningxia Medical University, Yinchuan, 750004, China.
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
20
|
Nerín-Fonz F, Cournia Z. Machine learning approaches in predicting allosteric sites. Curr Opin Struct Biol 2024; 85:102774. [PMID: 38354652 DOI: 10.1016/j.sbi.2024.102774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/29/2023] [Accepted: 01/02/2024] [Indexed: 02/16/2024]
Abstract
Allosteric regulation is a fundamental biological mechanism that can control critical cellular processes via allosteric modulator binding to protein distal functional sites. The advantages of allosteric modulators over orthosteric ones have sparked the development of numerous computational approaches, such as the identification of allosteric binding sites, to facilitate allosteric drug discovery. Building on the success of machine learning (ML) models for solving complex problems in biology and chemistry, several ML models for predicting allosteric sites have been developed. In this review, we provide an overview of these models and discuss future perspectives powered by the field of artificial intelligence such as protein language models.
Collapse
Affiliation(s)
- Francho Nerín-Fonz
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, Athens 11527, Greece; Vienna Doctoral School of Pharmaceutical, Nutritional and Sport Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
| | - Zoe Cournia
- Biomedical Research Foundation, Academy of Athens, 4 Soranou Ephesiou, Athens 11527, Greece; Vienna Doctoral School of Pharmaceutical, Nutritional and Sport Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
| |
Collapse
|
21
|
Martinez-Corral R, Nam KM, DePace AH, Gunawardena J. The Hill function is the universal Hopfield barrier for sharpness of input-output responses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.27.587054. [PMID: 38585761 PMCID: PMC10996692 DOI: 10.1101/2024.03.27.587054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The Hill functions, ℋ h ( x ) = x h / 1 + x h , have been widely used in biology for over a century but, with the exception of ℋ 1 , they have had no justification other than as a convenient fit to empirical data. Here, we show that they are the universal limit for the sharpness of any input-output response arising from a Markov process model at thermodynamic equilibrium. Models may represent arbitrary molecular complexity, with multiple ligands, internal states, conformations, co-regulators, etc, under core assumptions that are detailed in the paper. The model output may be any linear combination of steady-state probabilities, with components other than the chosen input ligand held constant. This formulation generalises most of the responses in the literature. We use a coarse-graining method in the graph-theoretic linear framework to show that two sharpness measures for input-output responses fall within an effectively bounded region of the positive quadrant, Ω m ⊂ ℝ + 2 , for any equilibrium model with m input binding sites. Ω m exhibits a cusp which approaches, but never exceeds, the sharpness of ℋ m but the region and the cusp can be exceeded when models are taken away from thermodynamic equilibrium. Such fundamental thermodynamic limits are called Hopfield barriers and our results provide a biophysical justification for the Hill functions as the universal Hopfield barriers for sharpness. Our results also introduce an object, Ω m , whose structure may be of mathematical interest, and suggest the importance of characterising Hopfield barriers for other forms of cellular information processing.
Collapse
Affiliation(s)
| | - Kee-Myoung Nam
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Angela H. DePace
- Howard Hughes Medical Institute, Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Jeremy Gunawardena
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
22
|
Habibpour M, Razaghi-Moghadam Z, Nikoloski Z. Prediction and integration of metabolite-protein interactions with genome-scale metabolic models. Metab Eng 2024; 82:216-224. [PMID: 38367764 DOI: 10.1016/j.ymben.2024.02.008] [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: 11/30/2023] [Revised: 01/13/2024] [Accepted: 02/14/2024] [Indexed: 02/19/2024]
Abstract
Metabolites, as small molecules, can act not only as substrates to enzymes, but also as effectors of activity of proteins with different functions, thereby affecting various cellular processes. While several experimental techniques have started to catalogue the metabolite-protein interactions (MPIs) present in different cellular contexts, characterizing the functional relevance of MPIs remains a challenging problem. Computational approaches from the constrained-based modeling framework allow for predicting MPIs and integrating their effects in the in silico analysis of metabolic and physiological phenotypes, like cell growth. Here, we provide a classification of all existing constraint-based approaches that predict and integrate MPIs using genome-scale metabolic networks as input. In addition, we benchmark the performance of the approaches to predict MPIs in a comparative study using different features extracted from the model structure and predicted metabolic phenotypes with the state-of-the-art metabolic networks of Escherichia coli and Saccharomyces cerevisiae. Lastly, we provide an outlook for future, feasible directions to expand the consideration of MPIs in constraint-based modeling approaches with wide biotechnological applications.
Collapse
Affiliation(s)
- Mahdis Habibpour
- Systems Biology and Mathematical Modeling Group, Max Planck Institute of Molecular Plant Physiology, 14476, Potsdam, Germany
| | - Zahra Razaghi-Moghadam
- Systems Biology and Mathematical Modeling Group, Max Planck Institute of Molecular Plant Physiology, 14476, Potsdam, Germany; Bioinformatics Department, Institute of Biochemistry and Biology, University of Potsdam, 14476, Potsdam, Germany
| | - Zoran Nikoloski
- Systems Biology and Mathematical Modeling Group, Max Planck Institute of Molecular Plant Physiology, 14476, Potsdam, Germany; Bioinformatics Department, Institute of Biochemistry and Biology, University of Potsdam, 14476, Potsdam, Germany.
| |
Collapse
|
23
|
Yaron JR, Bakkaloglu S, Grigaitis NA, Babur FH, Macko S, Rhodes S, Norvor-Davis S, Rege K. Inflammasome modulation with P2X7 inhibitor A438079-loaded dressings for diabetic wound healing. Front Immunol 2024; 15:1340405. [PMID: 38426101 PMCID: PMC10901979 DOI: 10.3389/fimmu.2024.1340405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
The inflammasome is a multiprotein complex critical for the innate immune response to injury. Inflammasome activation initiates healthy wound healing, but comorbidities with poor healing, including diabetes, exhibit pathologic, sustained activation with delayed resolution that prevents healing progression. In prior work, we reported the allosteric P2X7 antagonist A438079 inhibits extracellular ATP-evoked NLRP3 signaling by preventing ion flux, mitochondrial reactive oxygen species generation, NLRP3 assembly, mature IL-1β release, and pyroptosis. However, the short half-life in vivo limits clinical translation of this promising molecule. Here, we develop a controlled release scaffold to deliver A438079 as an inflammasome-modulating wound dressing for applications in poorly healing wounds. We fabricated and characterized tunable thickness, long-lasting silk fibroin dressings and evaluated A438079 loading and release kinetics. We characterized A438079-loaded silk dressings in vitro by measuring IL-1β release and inflammasome assembly by perinuclear ASC speck formation. We further evaluated the performance of A438079-loaded silk dressings in a full-thickness model of wound healing in genetically diabetic mice and observed acceleration of wound closure by 10 days post-wounding with reduced levels of IL-1β at the wound edge. This work provides a proof-of-principle for translating pharmacologic inhibition of ATP-induced inflammation in diabetic wounds and represents a novel approach to therapeutically targeting a dysregulated mechanism in diabetic wound impairment.
Collapse
Affiliation(s)
- Jordan R. Yaron
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
- School for Engineering of Matter, Transport & Energy, Arizona State University, Tempe, AZ, United States
| | - Selin Bakkaloglu
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Nicole A. Grigaitis
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
- Biological Design Graduate Program, Arizona State University, Tempe, AZ, United States
| | - Farhan H. Babur
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Sophia Macko
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Samantha Rhodes
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Solenne Norvor-Davis
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Kaushal Rege
- Center for Biomaterials Innovation and Translation, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
- School for Engineering of Matter, Transport & Energy, Arizona State University, Tempe, AZ, United States
- Biological Design Graduate Program, Arizona State University, Tempe, AZ, United States
- Chemical Engineering, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
24
|
He J, Liu X, Zhu C, Zha J, Li Q, Zhao M, Wei J, Li M, Wu C, Wang J, Jiao Y, Ning S, Zhou J, Hong Y, Liu Y, He H, Zhang M, Chen F, Li Y, He X, Wu J, Lu S, Song K, Lu X, Zhang J. ASD2023: towards the integrating landscapes of allosteric knowledgebase. Nucleic Acids Res 2024; 52:D376-D383. [PMID: 37870448 PMCID: PMC10767950 DOI: 10.1093/nar/gkad915] [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: 08/21/2023] [Revised: 09/22/2023] [Accepted: 10/06/2023] [Indexed: 10/24/2023] Open
Abstract
Allosteric regulation, induced by perturbations at an allosteric site topographically distinct from the orthosteric site, is one of the most direct and efficient ways to fine-tune macromolecular function. The Allosteric Database (ASD; accessible online at http://mdl.shsmu.edu.cn/ASD) has been systematically developed since 2009 to provide comprehensive information on allosteric regulation. In recent years, allostery has seen sustained growth and wide-ranging applications in life sciences, from basic research to new therapeutics development, while also elucidating emerging obstacles across allosteric research stages. To overcome these challenges and maintain high-quality data center services, novel features were curated in the ASD2023 update: (i) 66 589 potential allosteric sites, covering > 80% of the human proteome and constituting the human allosteric pocketome; (ii) 748 allosteric protein-protein interaction (PPI) modulators with clear mechanisms, aiding protein machine studies and PPI-targeted drug discovery; (iii) 'Allosteric Hit-to-Lead,' a pioneering dataset providing panoramic views from 87 well-defined allosteric hits to 6565 leads and (iv) 456 dualsteric modulators for exploring the simultaneous regulation of allosteric and orthosteric sites. Meanwhile, ASD2023 maintains a significant growth of foundational allosteric data. Based on these efforts, the allosteric knowledgebase is progressively evolving towards an integrated landscape, facilitating advancements in allosteric target identification, mechanistic exploration and drug discovery.
Collapse
Affiliation(s)
- Jixiao He
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xinyi Liu
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chunhao Zhu
- College of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, China
| | - Jinyin Zha
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Li
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Mingzhu Zhao
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiacheng Wei
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Mingyu Li
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chengwei Wu
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao-Tong University School of Medicine (SJTU-SM), Shanghai 200011, China
| | - Junyuan Wang
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao-Tong University School of Medicine (SJTU-SM), Shanghai 200011, China
| | - Yonglai Jiao
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shaobo Ning
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiamin Zhou
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao-Tong University School of Medicine (SJTU-SM), Shanghai 200011, China
| | - Yue Hong
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yonghui Liu
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hongxi He
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao-Tong University School of Medicine (SJTU-SM), Shanghai 200011, China
| | - Mingyang Zhang
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Feiying Chen
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yanxiu Li
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xinheng He
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jing Wu
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shaoyong Lu
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Kun Song
- Nutshell Therapeutics, Shanghai 201210, China
| | - Xuefeng Lu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao-Tong University School of Medicine (SJTU-SM), Shanghai 200011, China
| | - Jian Zhang
- State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medicinal Chemistry and Bioinformatics Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- College of Pharmacy, Ningxia Medical University, 1160 Shengli Street, Yinchuan, Ningxia 750004, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
25
|
Su J, Fu C, Wang S, Chen X, Wang R, Shi H, Li J, Wang X. Screening and Activity Evaluation of Novel BCR-ABL/T315I Tyrosine Kinase Inhibitors. Curr Med Chem 2024; 31:2872-2894. [PMID: 37211852 DOI: 10.2174/0929867330666230519105900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 04/03/2023] [Accepted: 04/13/2023] [Indexed: 05/23/2023]
Abstract
INTRODUCTION Chronic myeloid leukemia (CML) is a kind of malignant tumor formed by the clonal proliferation of bone marrow hematopoietic stem cells. BCR-ABL fusion protein, found in more than 90% of patients, is a vital target for discovering anti- CML drugs. Up to date, imatinib is the first BCR-ABL tyrosine kinase inhibitor (TKI) approved by the FDA for treating CML. However, the drug resistance problems appeared for many reasons, especially the T135I mutation, a "gatekeeper" of BCR-ABL. Currently, there is no long-term effective and low side effect drug in clinical. METHODS This study intends to find novel TKIs targeting BCR-ABL with high inhibitory activity against T315I mutant protein by combining artificial intelligence technology and cell growth curve, cytotoxicity, flow cytometry and Western blot experiments. RESULTS The obtained compound was found to kill leukemia cells, which had good inhibitory efficacy in BaF3/T315I cells. Compound no 4 could induce cell cycle arrest, cause autophagy and apoptosis, and inhibit the phosphorylation of BCR-ABL tyrosine kinase, STAT5 and Crkl proteins. CONCLUSION The results indicated that the screened compound could be used as a lead compound for further research to discover ideal chronic myeloid leukemia therapeutic drugs.
Collapse
MESH Headings
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/metabolism
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/chemistry
- Humans
- Apoptosis/drug effects
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Cell Proliferation/drug effects
- Drug Screening Assays, Antitumor
- Cell Line, Tumor
- Mice
- Animals
- Autophagy/drug effects
- STAT5 Transcription Factor/metabolism
- STAT5 Transcription Factor/antagonists & inhibitors
- Cell Cycle Checkpoints/drug effects
- Tyrosine Kinase Inhibitors
- Adaptor Proteins, Signal Transducing
Collapse
Affiliation(s)
- Jie Su
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., 730000, Lanzhou, China
| | - Chenggong Fu
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., 730000, Lanzhou, China
| | - Shuo Wang
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., 730000, Lanzhou, China
| | - Xuelian Chen
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., 730000, Lanzhou, China
| | - Runan Wang
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., 730000, Lanzhou, China
| | - Huaihuai Shi
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., 730000, Lanzhou, China
| | - Jiazhong Li
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., 730000, Lanzhou, China
| | - Xin Wang
- School of Pharmacy, Lanzhou University, 199 West Donggang Rd., 730000, Lanzhou, China
| |
Collapse
|
26
|
Chen Y, Xiao L, Qiu J. Neuronomodulation of Excitable Neurons. Neurosci Bull 2024; 40:103-112. [PMID: 37584858 PMCID: PMC10774251 DOI: 10.1007/s12264-023-01095-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 06/03/2023] [Indexed: 08/17/2023] Open
Abstract
Neuronomodulation refers to the modulation of neural conduction and synaptic transmission (i.e., the conduction process involved in synaptic transmission) of excitable neurons via changes in the membrane potential in response to chemical substances, from spillover neurotransmitters to paracrine or endocrine hormones circulating in the blood. Neuronomodulation can be direct or indirect, depending on the transduction pathways from the ligand binding site to the ion pore, either on the same molecule, i.e. the ion channel, or through an intermediate step on different molecules. The major players in direct neuronomodulation are ligand-gated or voltage-gated ion channels. The key process of direct neuronomodulation is the binding and chemoactivation of ligand-gated or voltage-gated ion channels, either orthosterically or allosterically, by various ligands. Indirect neuronomodulation involves metabotropic receptor-mediated slow potentials, where steroid hormones, cytokines, and chemokines can implement these actions. Elucidating neuronomodulation is of great significance for understanding the physiological mechanisms of brain function, and the occurrence and treatment of diseases.
Collapse
Affiliation(s)
- Yizhang Chen
- Institute of Neuroscience, Second Military Medical University, Shanghai, 200433, China.
| | - Lin Xiao
- Institute for Brain Research and Rehabilitation, Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, South China Normal University, Guangzhou, 510631, China.
| | - Jian Qiu
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, 97239, USA.
| |
Collapse
|
27
|
Bao Y, Xu Y, Jia F, Li M, Xu R, Zhang F, Guo J. Allosteric inhibition of myosin by phenamacril: a synergistic mechanism revealed by computational and experimental approaches. PEST MANAGEMENT SCIENCE 2023; 79:4977-4989. [PMID: 37540764 DOI: 10.1002/ps.7699] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/29/2023] [Accepted: 08/04/2023] [Indexed: 08/06/2023]
Abstract
BACKGROUND Myosin plays a crucial role in cellular processes, while its dysfunction can lead to organismal malfunction. Phenamacril (PHA), a highly species-specific and non-competitive inhibitor of myosin I (FgMyoI) from Fusarium graminearum, has been identified as an effective fungicide for controlling plant diseases caused by partial Fusarium pathogens, such as wheat scab and rice bakanae. However, the molecular basis of its action is still unclear. RESULTS This study used multiple computational approaches first to elucidate the allosteric inhibition mechanism of FgMyoI by PHA at the atomistic level. The results indicated the increase of adenosine triphosphate (ATP) binding affinity upon PHA binding, which might impede the release of hydrolysis products. Furthermore, simulations revealed a broadened outer cleft and a significantly more flexible interface for actin binding, accompanied by a decrease in signaling transduction from the catalytic center to the actin-binding interface. These various effects might work together to disrupt the actomyosin cycle and hinder the ability of motor to generate force. Our experimental results further confirmed that PHA reduces the enzymatic activity of myosin and its binding with actin. CONCLUSION Therefore, our findings demonstrated that PHA might suppress the function of myosin through a synergistic mechanism, providing new insights into myosin allostery and offering new avenues for drug/fungicide discovery targeting myosin. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yiqiong Bao
- College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Yan Xu
- College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Fangying Jia
- College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Mengrong Li
- College of Life Sciences, Nanjing Agricultural University, Nanjing, China
| | - Ran Xu
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao, China
| | - Feng Zhang
- College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Jingjing Guo
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao, China
- Engineering Research Centre of Applied Technology on Machine Translation and Artificial Intelligence, Macao Polytechnic University, Macao, China
| |
Collapse
|
28
|
Do HN, Wang J, Miao Y. Deep Learning Dynamic Allostery of G-Protein-Coupled Receptors. JACS AU 2023; 3:3165-3180. [PMID: 38034960 PMCID: PMC10685416 DOI: 10.1021/jacsau.3c00503] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/10/2023] [Accepted: 10/18/2023] [Indexed: 12/02/2023]
Abstract
G-protein-coupled receptors (GPCRs) make up the largest superfamily of human membrane proteins and represent primary targets of ∼1/3 of currently marketed drugs. Allosteric modulators have emerged as more selective drug candidates compared with orthosteric agonists and antagonists. However, many X-ray and cryo-EM structures of GPCRs resolved so far exhibit negligible differences upon the binding of positive and negative allosteric modulators (PAMs and NAMs). The mechanism of dynamic allosteric modulation in GPCRs remains unclear. In this work, we have systematically mapped dynamic changes in free energy landscapes of GPCRs upon binding of allosteric modulators using the Gaussian accelerated molecular dynamics (GaMD), deep learning (DL), and free energy prOfiling Workflow (GLOW). GaMD simulations were performed for a total of 66 μs on 44 GPCR systems in the presence and absence of the modulator. DL and free energy calculations revealed significantly reduced dynamic fluctuations and conformational space of GPCRs upon modulator binding. While the modulator-free GPCRs often sampled multiple low-energy conformational states, the NAMs and PAMs confined the inactive and active agonist-G-protein-bound GPCRs, respectively, to mostly only one specific conformation for signaling. Such cooperative effects were significantly reduced for binding of the selective modulators to "non-cognate" receptor subtypes. Therefore, GPCR allostery exhibits a dynamic "conformational selection" mechanism. In the absence of available modulator-bound structures as for most current GPCRs, it is critical to use a structural ensemble of representative GPCR conformations rather than a single structure for compound docking ("ensemble docking"), which will potentially improve structure-based design of novel allosteric drugs of GPCRs.
Collapse
Affiliation(s)
| | - Jinan Wang
- Computational Biology Program
and Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047, United States
| | | |
Collapse
|
29
|
Nussinov R, Liu Y, Zhang W, Jang H. Protein conformational ensembles in function: roles and mechanisms. RSC Chem Biol 2023; 4:850-864. [PMID: 37920394 PMCID: PMC10619138 DOI: 10.1039/d3cb00114h] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/02/2023] [Indexed: 11/04/2023] Open
Abstract
The sequence-structure-function paradigm has dominated twentieth century molecular biology. The paradigm tacitly stipulated that for each sequence there exists a single, well-organized protein structure. Yet, to sustain cell life, function requires (i) that there be more than a single structure, (ii) that there be switching between the structures, and (iii) that the structures be incompletely organized. These fundamental tenets called for an updated sequence-conformational ensemble-function paradigm. The powerful energy landscape idea, which is the foundation of modernized molecular biology, imported the conformational ensemble framework from physics and chemistry. This framework embraces the recognition that proteins are dynamic and are always interconverting between conformational states with varying energies. The more stable the conformation the more populated it is. The changes in the populations of the states are required for cell life. As an example, in vivo, under physiological conditions, wild type kinases commonly populate their more stable "closed", inactive, conformations. However, there are minor populations of the "open", ligand-free states. Upon their stabilization, e.g., by high affinity interactions or mutations, their ensembles shift to occupy the active states. Here we discuss the role of conformational propensities in function. We provide multiple examples of diverse systems, including protein kinases, lipid kinases, and Ras GTPases, discuss diverse conformational mechanisms, and provide a broad outlook on protein ensembles in the cell. We propose that the number of molecules in the active state (inactive for repressors), determine protein function, and that the dynamic, relative conformational propensities, rather than the rigid structures, are the hallmark of cell life.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research Frederick MD 21702 USA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University Tel Aviv 69978 Israel
- Cancer Innovation Laboratory, National Cancer Institute Frederick MD 21702 USA
| | - Yonglan Liu
- Cancer Innovation Laboratory, National Cancer Institute Frederick MD 21702 USA
| | - Wengang Zhang
- Cancer Innovation Laboratory, National Cancer Institute Frederick MD 21702 USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research Frederick MD 21702 USA
- Cancer Innovation Laboratory, National Cancer Institute Frederick MD 21702 USA
| |
Collapse
|
30
|
Djemili R, Adrouche S, Durot S, Heitz V. Allosterically Driven Assembly of a Multisite Cage-Based [2]Semirotaxane. J Org Chem 2023; 88:14760-14766. [PMID: 37812736 DOI: 10.1021/acs.joc.3c01381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
The assembly of a [2]semirotaxane from a half-dumbbell endowed with a pyrazine coordination site and a bis-Zn(II) porphyrin cage as a multisite ring is reported. The threading is allosterically driven by the coordination of silver(I) ions to the multiple binding sites of the cage linkers, as shown by NMR studies. Addition of chloride ions destabilizes [2]semirotaxane, leading to its disassembly into its cage and half-dumbbell components.
Collapse
Affiliation(s)
- Ryan Djemili
- Laboratoire de Synthèse des Assemblages Moléculaires Multifonctionnels Institut de Chimie de Strasbourg, CNRS/UMR 7177, Université de Strasbourg, 4 rue Blaise Pascal, 67000 Strasbourg, France
| | - Sonia Adrouche
- Laboratoire de Synthèse des Assemblages Moléculaires Multifonctionnels Institut de Chimie de Strasbourg, CNRS/UMR 7177, Université de Strasbourg, 4 rue Blaise Pascal, 67000 Strasbourg, France
| | - Stéphanie Durot
- Laboratoire de Synthèse des Assemblages Moléculaires Multifonctionnels Institut de Chimie de Strasbourg, CNRS/UMR 7177, Université de Strasbourg, 4 rue Blaise Pascal, 67000 Strasbourg, France
| | - Valérie Heitz
- Laboratoire de Synthèse des Assemblages Moléculaires Multifonctionnels Institut de Chimie de Strasbourg, CNRS/UMR 7177, Université de Strasbourg, 4 rue Blaise Pascal, 67000 Strasbourg, France
| |
Collapse
|
31
|
Di Marino D, Conflitti P, Motta S, Limongelli V. Structural basis of dimerization of chemokine receptors CCR5 and CXCR4. Nat Commun 2023; 14:6439. [PMID: 37833254 PMCID: PMC10575954 DOI: 10.1038/s41467-023-42082-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are prominent drug targets responsible for extracellular-to-intracellular signal transduction. GPCRs can form functional dimers that have been poorly characterized so far. Here, we show the dimerization mechanism of the chemokine receptors CCR5 and CXCR4 by means of an advanced free-energy technique named coarse-grained metadynamics. Our results reproduce binding events between the GPCRs occurring in the minute timescale, revealing a symmetric and an asymmetric dimeric structure for each of the three investigated systems, CCR5/CCR5, CXCR4/CXCR4, and CCR5/CXCR4. The transmembrane helices TM4-TM5 and TM6-TM7 are the preferred binding interfaces for CCR5 and CXCR4, respectively. The identified dimeric states differ in the access to the binding sites of the ligand and G protein, indicating that dimerization may represent a fine allosteric mechanism to regulate receptor activity. Our study offers structural basis for the design of ligands able to modulate the formation of CCR5 and CXCR4 dimers and in turn their activity, with therapeutic potential against HIV, cancer, and immune-inflammatory diseases.
Collapse
Affiliation(s)
- Daniele Di Marino
- Department of Life and Environmental Sciences - New York-Marche Structural Biology Centre (NY-MaSBiC), Polytechnic University of Marche, Via Brecce Bianche, 60131, Ancona, Italy
- Neuronal Death and Neuroprotection Unit, Department of Neuroscience, Mario Negri Institute for Pharmacological Research-IRCCS, Via Mario Negri 2, 20156, Milan, Italy
- National Biodiversity Future Center (NBFC), Palermo, Italy
| | - Paolo Conflitti
- Università della Svizzera italiana (USI), Faculty of Biomedical Sciences, Euler Institute, Via G. Buffi 13, CH-6900, Lugano, Switzerland
| | - Stefano Motta
- Department of Earth and Environmental Sciences, University of Milano-Bicocca, Piazza della Scienza 1, 20126, Milan, Italy
| | - Vittorio Limongelli
- Università della Svizzera italiana (USI), Faculty of Biomedical Sciences, Euler Institute, Via G. Buffi 13, CH-6900, Lugano, Switzerland.
| |
Collapse
|
32
|
Yang L, Zhu X, Finlay DB, Green H, Glass M, Duffull SB. A kinetic model for positive allosteric modulator (PAM)-antagonists for the type 1 cannabinoid (CB 1 ) receptor. Br J Pharmacol 2023; 180:2661-2676. [PMID: 37277184 DOI: 10.1111/bph.16158] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 03/29/2023] [Accepted: 05/22/2023] [Indexed: 06/07/2023] Open
Abstract
BACKGROUND AND PURPOSE The cannabinoid (CB1 ) receptor is among the most abundant G protein-coupled receptors in brain. Allosteric ligands bind to a different site on receptors than the orthosteric ligand can have effects that are unique to the allosteric ligand and modulate orthosteric ligand activity. We propose a unified mathematical model describing the interaction effects of the allosteric ligand Org27569 and the orthosteric agonist CP55940 on CB1 receptor. EXPERIMENTAL APPROACH A ternary complex model was constructed, which incorporated kinetic properties to describe the time course of effects of Org27569 and CP55940 reported in the literature: (i) enhanced receptor binding of CP55940, (ii) reduced internalisation and (iii), time-dependent modulation of cAMP. Underlying mechanisms of time-dependent modulation by Org27569 were evaluated by simulation. KEY RESULTS A hypothetical transitional state of CP55940-CB1 -Org27569, which can internalise but cannot inhibit cAMP, was shown to be necessary and was sufficient to describe the allosteric modulation by Org27569, prior to receptors adopting an inactive conformation. The model indicated that the formation of this transitional CP55940-CB1 -Org27569 state and final inactive CP55940-CB1 -Org27569 state contributes to the enhanced CP55940 binding. The inactive CP55940-CB1 -Org27569 cannot internalise or inhibit cAMP, leading to reduced internalisation and cessation of cAMP inhibition. CONCLUSIONS AND IMPLICATIONS In conclusion, a kinetic mathematical model for CB1 receptor allosteric modulation was developed. However, a standard ternary complex model was not sufficient to capture the data and a hypothetical transitional state was required to describe the allosteric modulation properties of Org27569.
Collapse
Affiliation(s)
- Liang Yang
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Otago Pharmacometrics Group, School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Xiao Zhu
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai, China
| | - David B Finlay
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Hayley Green
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Michelle Glass
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Stephen B Duffull
- Otago Pharmacometrics Group, School of Pharmacy, University of Otago, Dunedin, New Zealand
- Certara, Princeton, New Jersey, USA
| |
Collapse
|
33
|
Kolotyeva NA, Gilmiyarova FN, Averchuk AS, Baranich TI, Rozanova NA, Kukla MV, Tregub PP, Salmina AB. Novel Approaches to the Establishment of Local Microenvironment from Resorbable Biomaterials in the Brain In Vitro Models. Int J Mol Sci 2023; 24:14709. [PMID: 37834155 PMCID: PMC10572431 DOI: 10.3390/ijms241914709] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/19/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
The development of brain in vitro models requires the application of novel biocompatible materials and biopolymers as scaffolds for controllable and effective cell growth and functioning. The "ideal" brain in vitro model should demonstrate the principal features of brain plasticity like synaptic transmission and remodeling, neurogenesis and angiogenesis, and changes in the metabolism associated with the establishment of new intercellular connections. Therefore, the extracellular scaffolds that are helpful in the establishment and maintenance of local microenvironments supporting brain plasticity mechanisms are of critical importance. In this review, we will focus on some carbohydrate metabolites-lactate, pyruvate, oxaloacetate, malate-that greatly contribute to the regulation of cell-to-cell communications and metabolic plasticity of brain cells and on some resorbable biopolymers that may reproduce the local microenvironment enriched in particular cell metabolites.
Collapse
Affiliation(s)
| | - Frida N. Gilmiyarova
- Department of Fundamental and Clinical Biochemistry with Laboratory Diagnostics, Samara State Medical University, 443099 Samara, Russia
| | - Anton S. Averchuk
- Brain Science Institute, Research Center of Neurology, 125367 Moscow, Russia
| | - Tatiana I. Baranich
- Brain Science Institute, Research Center of Neurology, 125367 Moscow, Russia
| | | | - Maria V. Kukla
- Brain Science Institute, Research Center of Neurology, 125367 Moscow, Russia
| | - Pavel P. Tregub
- Brain Science Institute, Research Center of Neurology, 125367 Moscow, Russia
- Department of Pathophysiology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Alla B. Salmina
- Brain Science Institute, Research Center of Neurology, 125367 Moscow, Russia
| |
Collapse
|
34
|
Schlessinger A, Zatorski N, Hutchinson K, Colas C. Targeting SLC transporters: small molecules as modulators and therapeutic opportunities. Trends Biochem Sci 2023; 48:801-814. [PMID: 37355450 PMCID: PMC10525040 DOI: 10.1016/j.tibs.2023.05.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/26/2023]
Abstract
Solute carrier (SLCs) transporters mediate the transport of a broad range of solutes across biological membranes. Dysregulation of SLCs has been associated with various pathologies, including metabolic and neurological disorders, as well as cancer and rare diseases. SLCs are therefore emerging as key targets for therapeutic intervention with several recently approved drugs targeting these proteins. Unlocking this large and complex group of proteins is essential to identifying unknown SLC targets and developing next-generation SLC therapeutics. Recent progress in experimental and computational techniques has significantly advanced SLC research, including drug discovery. Here, we review emerging topics in therapeutic discovery of SLCs, focusing on state-of-the-art approaches in structural, chemical, and computational biology, and discuss current challenges in transporter drug discovery.
Collapse
Affiliation(s)
- Avner Schlessinger
- Department of Pharmacological Sciences Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Nicole Zatorski
- Department of Pharmacological Sciences Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Keino Hutchinson
- Department of Pharmacological Sciences Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Claire Colas
- University of Vienna, Department of Pharmaceutical Chemistry, Vienna, Austria.
| |
Collapse
|
35
|
Flatt S, Busiello DM, Zamuner S, De Los Rios P. ABC transporters are billion-year-old Maxwell Demons. COMMUNICATIONS PHYSICS 2023; 6:205. [PMID: 38665399 PMCID: PMC11041718 DOI: 10.1038/s42005-023-01320-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/26/2023] [Indexed: 04/28/2024]
Abstract
ATP-Binding Cassette (ABC) transporters are a broad family of biological machines, found in most prokaryotic and eukaryotic cells, performing the crucial import or export of substrates through both plasma and organellar membranes, and maintaining a steady concentration gradient driven by ATP hydrolysis. Building upon the present biophysical and biochemical characterization of ABC transporters, we propose here a model whose solution reveals that these machines are an exact molecular realization of the autonomous Maxwell Demon, a century-old abstract device that uses an energy source to drive systems away from thermodynamic equilibrium. In particular, the Maxwell Demon does not perform any direct mechanical work on the system, but simply selects which spontaneous processes to allow and which ones to forbid based on information that it collects and processes. In its autonomous version, the measurement device is embedded in the system itself. In the molecular model introduced here, the different operations that characterize Maxwell Demons (measurement, feedback, resetting) are features that emerge from the biochemical and structural properties of ABC transporters, revealing the crucial role of allostery to process information. Our framework allows us to develop an explicit bridge between the molecular-level description and the higher-level language of information theory for ABC transporters.
Collapse
Affiliation(s)
- Solange Flatt
- Institute of Physics, School of Basic Sciences, École Polytechnique Fédérale de Lausanne—EPFL, Lausanne, 1015 Switzerland
| | - Daniel Maria Busiello
- Institute of Physics, School of Basic Sciences, École Polytechnique Fédérale de Lausanne—EPFL, Lausanne, 1015 Switzerland
- Max Planck Institute for the Physics of Complex Systems, Dresden, 01187 Germany
| | - Stefano Zamuner
- Institute of Physics, School of Basic Sciences, École Polytechnique Fédérale de Lausanne—EPFL, Lausanne, 1015 Switzerland
| | - Paolo De Los Rios
- Institute of Physics, School of Basic Sciences, École Polytechnique Fédérale de Lausanne—EPFL, Lausanne, 1015 Switzerland
- Institute of Bioengineering, School of Life Sciences, École Polytechnique Fédérale de Lausanne—EPFL, Lausanne, 1015 Switzerland
| |
Collapse
|
36
|
Bosquez-Berger T, Gudorf JA, Kuntz CP, Desmond JA, Schlebach JP, VanNieuwenhze MS, Straiker A. Structure-Activity Relationship Study of Cannabidiol-Based Analogs as Negative Allosteric Modulators of the μ-Opioid Receptor. J Med Chem 2023; 66:9466-9494. [PMID: 37437224 PMCID: PMC11299522 DOI: 10.1021/acs.jmedchem.3c00061] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
The US faces an unprecedented surge in fatal drug overdoses. Naloxone, the only antidote for opiate overdose, competes at the mu opioid receptor (μOR) orthosteric site. Naloxone struggles against fentanyl-class synthetic opioids that now cause ∼80% of deaths. Negative allosteric modulators (NAMs) targeting secondary sites may noncompetitively downregulate μOR activation. (-)-Cannabidiol ((-)-CBD) is a candidate μOR NAM. To explore its therapeutic potential, we evaluated the structure-activity relationships among CBD analogs to identify NAMs with increased potency. Using a cyclic AMP assay, we characterize reversal of μOR activation by 15 CBD analogs, several of which proved more potent than (-)-CBD. Comparative docking investigations suggest that potent compounds interact with a putative allosteric pocket to stabilize the inactive μOR conformation. Finally, these compounds enhance naloxone displacement of fentanyl from the orthosteric site. Our results suggest that CBD analogs offer considerable potential for the development of next-generation antidotes for opioid overdose.
Collapse
Affiliation(s)
- Taryn Bosquez-Berger
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Program in Neuroscience, Indiana University, Bloomington, Indiana 47405, United States
| | - Jessica A Gudorf
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Charles P Kuntz
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Jacob A Desmond
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | - Jonathan P Schlebach
- Department of Chemistry, Indiana University, Bloomington, Indiana 47405, United States
| | | | - Alex Straiker
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Program in Neuroscience, Indiana University, Bloomington, Indiana 47405, United States
| |
Collapse
|
37
|
Wang S, Xie J, Pei J, Lai L. CavityPlus 2022 Update: An Integrated Platform for Comprehensive Protein Cavity Detection and Property Analyses with User-friendly Tools and Cavity Databases. J Mol Biol 2023; 435:168141. [PMID: 37356903 DOI: 10.1016/j.jmb.2023.168141] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/13/2023] [Accepted: 04/27/2023] [Indexed: 06/27/2023]
Abstract
Ligand binding sites provide essential information for uncovering protein functions and structure-based drug discovery. To facilitate cavity detection and property analysis process, we developed a comprehensive web server, CavityPlus in 2018. CavityPlus applies the CAVITY program to detect potential binding sites in a given protein structure. The CavPharmer, CorrSite, and CovCys tools can then be applied to generate receptor-based pharmacophore models, identify potential allosteric sites, or detect druggable cysteine residues for covalent drug design. While CavityPlus has been widely used, the constantly evolving knowledge and methods make it necessary to improve and extend its functions. This study presents a new version of CavityPlus, CavityPlus 2022 through a series of upgrades. We upgraded the CAVITY tool to greatly speed up cavity detection calculation. We optimized the CavPharmer tool for fast speed and more accurate results. We integrated the newly developed CorrSite2.0 into the CavityPlus 2022 web server for its improved performance of allosteric site prediction. We also added a new CavityMatch module for drug repurposing and protein function studies by searching similar cavities to a given cavity from pre-constructed cavity databases. The new version of CavityPlus is freely available at http://pkumdl.cn:8000/cavityplus/.
Collapse
Affiliation(s)
- Shiwei Wang
- BMLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing, PR China
| | - Juan Xie
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, PR China
| | - Jianfeng Pei
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, PR China; Research Unit of Drug Design Method, Chinese Academy of Medical Sciences (2021RU014), Beijing 100871, PR China
| | - Luhua Lai
- BMLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing, PR China; Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, PR China; Research Unit of Drug Design Method, Chinese Academy of Medical Sciences (2021RU014), Beijing 100871, PR China.
| |
Collapse
|
38
|
Dvorak V, Superti-Furga G. Structural and functional annotation of solute carrier transporters: implication for drug discovery. Expert Opin Drug Discov 2023; 18:1099-1115. [PMID: 37563933 DOI: 10.1080/17460441.2023.2244760] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/01/2023] [Indexed: 08/12/2023]
Abstract
INTRODUCTION Solute carriers (SLCs) represent the largest group of membrane transporters in the human genome. They play a central role in controlling the compartmentalization of metabolism and most of this superfamily is linked to human disease. Despite being in general considered druggable and attractive therapeutic targets, many SLCs remain poorly annotated, both functionally and structurally. AREAS COVERED The aim of this review is to provide an overview of functional and structural parameters of SLCs that play important roles in their druggability. To do this, the authors provide an overview of experimentally solved structures of human SLCs, with emphasis on structures solved in complex with chemical modulators. From the functional annotations, the authors focus on SLC localization and SLC substrate annotations. EXPERT OPINION Recent progress in the structural and functional annotations allows to refine the SLC druggability index. Particularly the increasing number of experimentally solved structures of SLCs provides insights into mode-of-action of a significant number of chemical modulators of SLCs.
Collapse
Affiliation(s)
- Vojtech Dvorak
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
39
|
Vuckovic Z, Wang J, Pham V, Mobbs JI, Belousoff MJ, Bhattarai A, Burger WAC, Thompson G, Yeasmin M, Nawaratne V, Leach K, van der Westhuizen ET, Khajehali E, Liang YL, Glukhova A, Wootten D, Lindsley CW, Tobin A, Sexton P, Danev R, Valant C, Miao Y, Christopoulos A, Thal DM. Pharmacological hallmarks of allostery at the M4 muscarinic receptor elucidated through structure and dynamics. eLife 2023; 12:83477. [PMID: 37248726 DOI: 10.7554/elife.83477] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 04/12/2023] [Indexed: 05/31/2023] Open
Abstract
Allosteric modulation of G protein-coupled receptors (GPCRs) is a major paradigm in drug discovery. Despite decades of research, a molecular-level understanding of the general principles that govern the myriad pharmacological effects exerted by GPCR allosteric modulators remains limited. The M4 muscarinic acetylcholine receptor (M4 mAChR) is a validated and clinically relevant allosteric drug target for several major psychiatric and cognitive disorders. In this study, we rigorously quantified the affinity, efficacy, and magnitude of modulation of two different positive allosteric modulators, LY2033298 (LY298) and VU0467154 (VU154), combined with the endogenous agonist acetylcholine (ACh) or the high-affinity agonist iperoxo (Ipx), at the human M4 mAChR. By determining the cryo-electron microscopy structures of the M4 mAChR, bound to a cognate Gi1 protein and in complex with ACh, Ipx, LY298-Ipx, and VU154-Ipx, and applying molecular dynamics simulations, we determine key molecular mechanisms underlying allosteric pharmacology. In addition to delineating the contribution of spatially distinct binding sites on observed pharmacology, our findings also revealed a vital role for orthosteric and allosteric ligand-receptor-transducer complex stability, mediated by conformational dynamics between these sites, in the ultimate determination of affinity, efficacy, cooperativity, probe dependence, and species variability. There results provide a holistic framework for further GPCR mechanistic studies and can aid in the discovery and design of future allosteric drugs.
Collapse
Affiliation(s)
- Ziva Vuckovic
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Jinan Wang
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, United States
| | - Vi Pham
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Jesse I Mobbs
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Matthew J Belousoff
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Apurba Bhattarai
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, United States
| | - Wessel A C Burger
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Geoff Thompson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Mahmuda Yeasmin
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Vindhya Nawaratne
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Katie Leach
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Emma T van der Westhuizen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Elham Khajehali
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Yi-Lynn Liang
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Alisa Glukhova
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Denise Wootten
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Craig W Lindsley
- Department of Pharmacology, Warren Center for Neuroscience Drug Discovery and Department of Chemistry, Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, United States
| | - Andrew Tobin
- The Centre for Translational Pharmacology, Advanced Research Centre (ARC), College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Patrick Sexton
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Radostin Danev
- Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Celine Valant
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Yinglong Miao
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, United States
| | - Arthur Christopoulos
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- Neuromedicines Discovery Centre, Monash University, Parkville, Australia
| | - David M Thal
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
- ARC Centre for Cryo-electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| |
Collapse
|
40
|
Ferré G, Gomes AAS, Louet M, Damian M, Bisch PM, Saurel O, Floquet N, Milon A, Banères JL. Sodium is a negative allosteric regulator of the ghrelin receptor. Cell Rep 2023; 42:112320. [PMID: 37027306 DOI: 10.1016/j.celrep.2023.112320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 11/09/2022] [Accepted: 03/14/2023] [Indexed: 04/08/2023] Open
Abstract
The functional properties of G protein-coupled receptors (GPCRs) are intimately associated with the different components in their cellular environment. Among them, sodium ions have been proposed to play a substantial role as endogenous allosteric modulators of GPCR-mediated signaling. However, this sodium effect and the underlying mechanisms are still unclear for most GPCRs. Here, we identified sodium as a negative allosteric modulator of the ghrelin receptor GHSR (growth hormone secretagogue receptor). Combining 23Na-nuclear magnetic resonance (NMR), molecular dynamics, and mutagenesis, we provide evidence that, in GHSR, sodium binds to the allosteric site conserved in class A GPCRs. We further leveraged spectroscopic and functional assays to show that sodium binding shifts the conformational equilibrium toward the GHSR-inactive ensemble, thereby decreasing basal and agonist-induced receptor-catalyzed G protein activation. All together, these data point to sodium as an allosteric modulator of GHSR, making this ion an integral component of the ghrelin signaling machinery.
Collapse
Affiliation(s)
- Guillaume Ferré
- Institut de Pharmacologie et de Biologie Structurale IPBS, Université de Toulouse UPS, CNRS, Toulouse, France
| | - Antoniel A S Gomes
- Institut des Biomolécules Max Mousseron IBMM, UMR-5247, University Montpellier, CNRS, ENSCM, Montpellier, France; Laboratório de Física Biológica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, Brazil
| | - Maxime Louet
- Institut des Biomolécules Max Mousseron IBMM, UMR-5247, University Montpellier, CNRS, ENSCM, Montpellier, France
| | - Marjorie Damian
- Institut des Biomolécules Max Mousseron IBMM, UMR-5247, University Montpellier, CNRS, ENSCM, Montpellier, France
| | - Paulo M Bisch
- Laboratório de Física Biológica, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-590, Brazil
| | - Olivier Saurel
- Institut de Pharmacologie et de Biologie Structurale IPBS, Université de Toulouse UPS, CNRS, Toulouse, France
| | - Nicolas Floquet
- Institut des Biomolécules Max Mousseron IBMM, UMR-5247, University Montpellier, CNRS, ENSCM, Montpellier, France
| | - Alain Milon
- Institut de Pharmacologie et de Biologie Structurale IPBS, Université de Toulouse UPS, CNRS, Toulouse, France.
| | - Jean-Louis Banères
- Institut des Biomolécules Max Mousseron IBMM, UMR-5247, University Montpellier, CNRS, ENSCM, Montpellier, France.
| |
Collapse
|
41
|
Zhang Y, Chen R, Zhang D, Qi S, Liu Y. Metabolite interactions between host and microbiota during health and disease: Which feeds the other? Biomed Pharmacother 2023; 160:114295. [PMID: 36709600 DOI: 10.1016/j.biopha.2023.114295] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/20/2023] [Accepted: 01/20/2023] [Indexed: 01/30/2023] Open
Abstract
Metabolites produced by the host and microbiota play a crucial role in how human bodies develop and remain healthy. Most of these metabolites are produced by microbiota and hosts in the digestive tract. Metabolites in the gut have important roles in energy metabolism, cellular communication, and host immunity, among other physiological activities. Although numerous host metabolites, such as free fatty acids, amino acids, and vitamins, are found in the intestine, metabolites generated by gut microbiota are equally vital for intestinal homeostasis. Furthermore, microbiota in the gut is the sole source of some metabolites, including short-chain fatty acids (SCFAs). Metabolites produced by microbiota, such as neurotransmitters and hormones, may modulate and significantly affect host metabolism. The gut microbiota is becoming recognized as a second endocrine system. A variety of chronic inflammatory disorders have been linked to aberrant host-microbiota interplays, but the precise mechanisms underpinning these disturbances and how they might lead to diseases remain to be fully elucidated. Microbiome-modulated metabolites are promising targets for new drug discovery due to their endocrine function in various complex disorders. In humans, metabolotherapy for the prevention or treatment of various disorders will be possible if we better understand the metabolic preferences of bacteria and the host in specific tissues and organs. Better disease treatments may be possible with the help of novel complementary therapies that target host or bacterial metabolism. The metabolites, their physiological consequences, and functional mechanisms of the host-microbiota interplays will be highlighted, summarized, and discussed in this overview.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Anethesiology, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China.
| | - Rui Chen
- Department of Pediatrics, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China.
| | - DuoDuo Zhang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, Jilin Province 130021, People's Republic of China.
| | - Shuang Qi
- Department of Anethesiology, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China.
| | - Yan Liu
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, People's Republic of China.
| |
Collapse
|
42
|
Katsu Y, Lin X, Ji R, Chen Z, Kamisaka Y, Bamba K, Baker ME. N-terminal domain influences steroid activation of the Atlantic sea lamprey corticoid receptor. J Steroid Biochem Mol Biol 2023; 228:106249. [PMID: 36646152 DOI: 10.1016/j.jsbmb.2023.106249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
Lampreys are jawless fish that evolved about 550 million years ago at the base of the vertebrate line. Modern lampreys contain a corticoid receptor (CR), the common ancestor of the glucocorticoid receptor (GR) and mineralocorticoid receptor (MR), which first appear in cartilaginous fish, such as sharks. Until recently, 344 amino acids at the amino terminus of adult lamprey CR were not present in the lamprey CR sequence in GenBank. A search of the recently sequenced lamprey germline genome identified two CR sequences, CR1 and CR2, containing the 344 previously un-identified amino acids. CR1 also contains a novel four amino acid insertion in the DNA-binding domain (DBD). We studied corticosteroid and progesterone activation of CR1 and CR2 and found their strongest response was to 11-deoxycorticosterone and 11-deoxycortisol, the two circulating corticosteroids in lamprey. Based on steroid specificity, both CRs are close to elephant shark MR and distant from elephant shark GR. HEK293 cells that were transfected with full-length CR1 or CR2 and the MMTV promoter have about 3-fold higher steroid-mediated activation compared to HEK293 cells transfected with these CRs and the TAT3 promoter. Deletion of the amino-terminal domain (NTD) of lamprey CR1 and CR2 to form truncated CRs decreased transcriptional activation by about 70% in HEK293 cells that were transfected with MMTV, but increased transcription by about 6-fold in cells transfected with TAT3. This indicated that the promoter has an important effect on NTD regulation of transcriptional activation of the CR by steroids. Our results also indicate that the entire lamprey CR sequence is needed for an accurate determination of steroid-mediated transcription.
Collapse
Affiliation(s)
| | - Xiaozhi Lin
- Graduate School of Life Science Hokkaido University Sapporo, Japan
| | - Ruigeng Ji
- Graduate School of Life Science Hokkaido University Sapporo, Japan
| | - Ze Chen
- Graduate School of Life Science Hokkaido University Sapporo, Japan
| | - Yui Kamisaka
- Graduate School of Life Science Hokkaido University Sapporo, Japan
| | - Koto Bamba
- Faculty of Science Hokkaido University Sapporo, Japan
| | - Michael E Baker
- Division of Nephrology-Hypertension Department of Medicine, 0693 University of California, San Diego 9500 Gilman Drive La Jolla, CA 92093-0693, USA; Center for Academic Research and Training in Anthropogeny (CARTA) University of California, San Diego La Jolla, CA 92093, USA.
| |
Collapse
|
43
|
Xie J, Pan G, Li Y, Lai L. How protein topology controls allosteric regulations. J Chem Phys 2023; 158:105102. [PMID: 36922138 DOI: 10.1063/5.0138279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
Abstract
Allostery is an important regulatory mechanism of protein functions. Among allosteric proteins, certain protein structure types are more observed. However, how allosteric regulation depends on protein topology remains elusive. In this study, we extracted protein topology graphs at the fold level and found that known allosteric proteins mainly contain multiple domains or subunits and allosteric sites reside more often between two or more domains of the same fold type. Only a small fraction of fold-fold combinations are observed in allosteric proteins, and homo-fold-fold combinations dominate. These analyses imply that the locations of allosteric sites including cryptic ones depend on protein topology. We further developed TopoAlloSite, a novel method that uses the kernel support vector machine to predict the location of allosteric sites on the overall protein topology based on the subgraph-matching kernel. TopoAlloSite successfully predicted known cryptic allosteric sites in several allosteric proteins like phosphopantothenoylcysteine synthetase, spermidine synthase, and sirtuin 6, demonstrating its power in identifying cryptic allosteric sites without performing long molecular dynamics simulations or large-scale experimental screening. Our study demonstrates that protein topology largely determines how its function can be allosterically regulated, which can be used to find new druggable targets and locate potential binding sites for rational allosteric drug design.
Collapse
Affiliation(s)
- Juan Xie
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Gaoxiang Pan
- BNLMS, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Yibo Li
- Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Luhua Lai
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| |
Collapse
|
44
|
Campana P, Nikoloski Z. Self- and cross-attention accurately predicts metabolite-protein interactions. NAR Genom Bioinform 2023; 5:lqad008. [PMID: 36733400 PMCID: PMC9887643 DOI: 10.1093/nargab/lqad008] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/20/2022] [Accepted: 01/17/2023] [Indexed: 02/04/2023] Open
Abstract
Metabolites regulate activity of proteins and thereby affect cellular processes in all organisms. Despite extensive efforts to catalogue the metabolite-protein interactome in different organisms by employing experimental and computational approaches, the coverage of such interactions remains fragmented, particularly for eukaryotes. Here, we make use of two most comprehensive collections, BioSnap and STITCH, of metabolite-protein interactions from seven eukaryotes as gold standards to train a deep learning model that relies on self- and cross-attention over protein sequences. This innovative protein-centric approach results in interaction-specific features derived from protein sequence alone. In addition, we designed and assessed a first double-blind evaluation protocol for metabolite-protein interactions, demonstrating the generalizability of the model. Our results indicated that the excellent performance of the proposed model over simpler alternatives and randomized baselines is due to the local and global features generated by the attention mechanisms. As a results, the predictions from the deep learning model provide a valuable resource for studying metabolite-protein interactions in eukaryotes.
Collapse
Affiliation(s)
- Pedro Alonso Campana
- Machine Learning, Department of Computer Science, University of Potsdam, 14476 Potsdam, Germany
- Bioinformatics, Institute of Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany
| | - Zoran Nikoloski
- Bioinformatics, Institute of Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany
- Systems Biology and Mathematical Modeling, Max Planck Institute of Molecular Plant Physiology, 14476 Potsdam, Germany
| |
Collapse
|
45
|
Do H, Wang J, Miao Y. Deep Learning Dynamic Allostery of G-Protein-Coupled Receptors. RESEARCH SQUARE 2023:rs.3.rs-2543463. [PMID: 36865316 PMCID: PMC9980202 DOI: 10.21203/rs.3.rs-2543463/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Abstract
G-protein-coupled receptors (GPCRs) are the largest superfamily of human membrane proteins and represent primary targets of ~ 1/3 of currently marketed drugs. Allosteric modulators have emerged as more selective drug candidates compared with orthosteric agonists and antagonists. However, many X-ray and cryo-EM structures of GPCRs resolved so far exhibit negligible differences upon binding of positive and negative allosteric modulators (PAMs and NAMs). Mechanism of dynamic allosteric modulation in GPCRs remains unclear. In this work, we have systematically mapped dynamic changes in free energy landscapes of GPCRs upon binding of allosteric modulators using the Gaussian accelerated molecular dynamics (GaMD), Deep Learning (DL) and free energy prOfiling Workflow (GLOW). A total of 18 available high-resolution experimental structures of allosteric modulator-bound class A and B GPCRs were collected for simulations. A number of 8 computational models were generated to examine selectivity of the modulators by changing their target receptors to different subtypes. All-atom GaMD simulations were performed for a total of 66 μs on 44 GPCR systems in the presence/absence of the modulator. DL and free energy calculations revealed significantly reduced conformational space of GPCRs upon modulator binding. While the modulator-free GPCRs often sampled multiple low-energy conformational states, the NAMs and PAMs confined the inactive and active agonist-G protein-bound GPCRs, respectively, to mostly only one specific conformation for signaling. Such cooperative effects were significantly reduced for binding of the selective modulators to "non-cognate" receptor subtypes in the computational models. Therefore, comprehensive DL of extensive GaMD simulations has revealed a general dynamic mechanism of GPCR allostery, which will greatly facilitate rational design of selective allosteric drugs of GPCRs.
Collapse
|
46
|
Xie J, Zhang W, Zhu X, Deng M, Lai L. Coevolution-based prediction of key allosteric residues for protein function regulation. eLife 2023; 12:81850. [PMID: 36799896 PMCID: PMC9981151 DOI: 10.7554/elife.81850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 02/16/2023] [Indexed: 02/18/2023] Open
Abstract
Allostery is fundamental to many biological processes. Due to the distant regulation nature, how allosteric mutations, modifications, and effector binding impact protein function is difficult to forecast. In protein engineering, remote mutations cannot be rationally designed without large-scale experimental screening. Allosteric drugs have raised much attention due to their high specificity and possibility of overcoming existing drug-resistant mutations. However, optimization of allosteric compounds remains challenging. Here, we developed a novel computational method KeyAlloSite to predict allosteric site and to identify key allosteric residues (allo-residues) based on the evolutionary coupling model. We found that protein allosteric sites are strongly coupled to orthosteric site compared to non-functional sites. We further inferred key allo-residues by pairwise comparing the difference of evolutionary coupling scores of each residue in the allosteric pocket with the functional site. Our predicted key allo-residues are in accordance with previous experimental studies for typical allosteric proteins like BCR-ABL1, Tar, and PDZ3, as well as key cancer mutations. We also showed that KeyAlloSite can be used to predict key allosteric residues distant from the catalytic site that are important for enzyme catalysis. Our study demonstrates that weak coevolutionary couplings contain important information of protein allosteric regulation function. KeyAlloSite can be applied in studying the evolution of protein allosteric regulation, designing and optimizing allosteric drugs, and performing functional protein design and enzyme engineering.
Collapse
Affiliation(s)
- Juan Xie
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijingChina
| | - Weilin Zhang
- BNLMS, Peking-Tsinghua Center for Life Sciences at the College of Chemistry and Molecular Engineering, Peking UniversityBeijingChina
| | - Xiaolei Zhu
- School of Sciences, Anhui Agricultural UniversityHefeiChina
| | - Minghua Deng
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijingChina
- School of Mathematical Sciences, Peking UniversityBeijingChina
- Center for Statistical Science, Peking UniversityBeijingChina
| | - Luhua Lai
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijingChina
- BNLMS, Peking-Tsinghua Center for Life Sciences at the College of Chemistry and Molecular Engineering, Peking UniversityBeijingChina
- Research Unit of Drug Design Method, Chinese Academy of Medical Sciences (2021RU014)BeijingChina
| |
Collapse
|
47
|
Shen S, Zhao C, Wu C, Sun S, Li Z, Yan W, Shao Z. Allosteric modulation of G protein-coupled receptor signaling. Front Endocrinol (Lausanne) 2023; 14:1137604. [PMID: 36875468 PMCID: PMC9978769 DOI: 10.3389/fendo.2023.1137604] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 01/31/2023] [Indexed: 02/18/2023] Open
Abstract
G protein-coupled receptors (GPCRs), the largest family of transmembrane proteins, regulate a wide array of physiological processes in response to extracellular signals. Although these receptors have proven to be the most successful class of drug targets, their complicated signal transduction pathways (including different effector G proteins and β-arrestins) and mediation by orthosteric ligands often cause difficulties for drug development, such as on- or off-target effects. Interestingly, identification of ligands that engage allosteric binding sites, which are different from classic orthosteric sites, can promote pathway-specific effects in cooperation with orthosteric ligands. Such pharmacological properties of allosteric modulators offer new strategies to design safer GPCR-targeted therapeutics for various diseases. Here, we explore recent structural studies of GPCRs bound to allosteric modulators. Our inspection of all GPCR families reveals recognition mechanisms of allosteric regulation. More importantly, this review highlights the diversity of allosteric sites and presents how allosteric modulators control specific GPCR pathways to provide opportunities for the development of new valuable agents.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei Yan
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
48
|
Do HN, Wang J, Miao Y. Deep Learning Dynamic Allostery of G-Protein-Coupled Receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.15.524128. [PMID: 36711515 PMCID: PMC9882226 DOI: 10.1101/2023.01.15.524128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
G-protein-coupled receptors (GPCRs) are the largest superfamily of human membrane proteins and represent primary targets of ~1/3 of currently marketed drugs. Allosteric modulators have emerged as more selective drug candidates compared with orthosteric agonists and antagonists. However, many X-ray and cryo-EM structures of GPCRs resolved so far exhibit negligible differences upon binding of positive and negative allosteric modulators (PAMs and NAMs). Mechanism of dynamic allosteric modulation in GPCRs remains unclear. In this work, we have systematically mapped dynamic changes in free energy landscapes of GPCRs upon binding of allosteric modulators using the Gaussian accelerated molecular dynamics (GaMD), Deep Learning (DL) and free energy prOfiling Workflow (GLOW). A total of 18 available high-resolution experimental structures of allosteric modulator-bound class A and B GPCRs were collected for simulations. A number of 8 computational models were generated to examine selectivity of the modulators by changing their target receptors to different subtypes. All-atom GaMD simulations were performed for a total of 66 μs on 44 GPCR systems in the presence/absence of the modulator. DL and free energy calculations revealed significantly reduced conformational space of GPCRs upon modulator binding. While the modulator-free GPCRs often sampled multiple low-energy conformational states, the NAMs and PAMs confined the inactive and active agonist-G protein-bound GPCRs, respectively, to mostly only one specific conformation for signaling. Such cooperative effects were significantly reduced for binding of the selective modulators to "non-cognate" receptor subtypes in the computational models. Therefore, comprehensive DL of extensive GaMD simulations has revealed a general dynamic mechanism of GPCR allostery, which will greatly facilitate rational design of selective allosteric drugs of GPCRs.
Collapse
Affiliation(s)
- Hung N. Do
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047
| | - Jinan Wang
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047
| | - Yinglong Miao
- Center for Computational Biology and Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas 66047
| |
Collapse
|
49
|
Cowgill J, Chanda B. Charge-voltage curves of Shaker potassium channel are not hysteretic at steady state. J Gen Physiol 2023; 155:213823. [PMID: 36692860 PMCID: PMC9884579 DOI: 10.1085/jgp.202112883] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/16/2022] [Accepted: 01/03/2023] [Indexed: 01/25/2023] Open
Abstract
Charge-voltage curves of many voltage-gated ion channels exhibit hysteresis but such curves are also a direct measure of free energy of channel gating and, hence, should be path-independent. Here, we identify conditions to measure steady-state charge-voltage curves and show that these are curves are not hysteretic. Charged residues in transmembrane segments of voltage-gated ion channels (VGICs) sense and respond to changes in the electric field. The movement of these gating charges underpins voltage-dependent activation and is also a direct metric of the net free-energy of channel activation. However, for most voltage-gated ion channels, the charge-voltage (Q-V) curves appear to be dependent on initial conditions. For instance, Q-V curves of Shaker potassium channel obtained by hyperpolarizing from 0 mV is left-shifted compared to those obtained by depolarizing from a holding potential of -80 mV. This hysteresis in Q-V curves is a common feature of channels in the VGIC superfamily and raises profound questions about channel energetics because the net free-energy of channel gating is a state function and should be path independent. Due to technical limitations, conventional gating current protocols are limited to test pulse durations of <500 ms, which raises the possibility that the dependence of Q-V on initial conditions reflects a lack of equilibration. Others have suggested that the hysteresis is fundamental thermodynamic property of voltage-gated ion channels and reflects energy dissipation due to measurements under non-equilibrium conditions inherent to rapid voltage jumps (Villalba-Galea. 2017. Channels. https://doi.org/10.1080/19336950.2016.1243190). Using an improved gating current and voltage-clamp fluorometry protocols, we show that the gating hysteresis arising from different initial conditions in Shaker potassium channel is eliminated with ultra-long (18-25 s) test pulses. Our study identifies a modified gating current recording protocol to obtain steady-state Q-V curves of a voltage-gated ion channel. Above all, these findings demonstrate that the gating hysteresis in Shaker channel is a kinetic phenomenon rather than a true thermodynamic property of the channel and the charge-voltage curve is a true measure of the net-free energy of channel gating.
Collapse
Affiliation(s)
- John Cowgill
- Departments of Anesthesiology, Neuroscience, Biochemistry and Molecular Biophysics, Center for Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO, USA,John Cowgill:
| | - Baron Chanda
- Departments of Anesthesiology, Neuroscience, Biochemistry and Molecular Biophysics, Center for Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO, USA,Correspondence to Baron Chanda:
| |
Collapse
|
50
|
Waclawiková B, Cesar Telles de Souza P, Schwalbe M, Neochoritis CG, Hoornenborg W, Nelemans SA, Marrink SJ, El Aidy S. Potential binding modes of the gut bacterial metabolite, 5-hydroxyindole, to the intestinal L-type calcium channels and its impact on the microbiota in rats. Gut Microbes 2023; 15:2154544. [PMID: 36511640 PMCID: PMC9754111 DOI: 10.1080/19490976.2022.2154544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Intestinal microbiota and microbiota-derived metabolites play a key role in regulating the host physiology. Recently, we have identified a gut-bacterial metabolite, namely 5-hydroxyindole, as a potent stimulant of intestinal motility via its modulation of L-type voltage-gated calcium channels located on the intestinal smooth muscle cells. Dysregulation of L-type voltage-gated calcium channels is associated with various gastrointestinal motility disorders, including constipation, making L-type voltage-gated calcium channels an important target for drug development. Nonetheless, the majority of currently available drugs are associated with alteration of the gut microbiota. Using 16S rRNA sequencing this study shows that, when administered orally, 5-hydroxyindole has only marginal effects on the rat cecal microbiota. Molecular dynamics simulations propose potential-binding pockets of 5-hydroxyindole in the α1 subunit of the L-type voltage-gated calcium channels and when its stimulatory effect on the rat colonic contractility was compared to 16 different analogues, ex-vivo, 5-hydroxyindole stood as the most potent enhancer of the intestinal contractility. Overall, the present findings imply a potential role of microbiota-derived metabolites as candidate therapeutics for targeted treatment of slow intestinal motility-related disorders including constipation.
Collapse
Affiliation(s)
- Barbora Waclawiková
- Host-Microbe Metabolic Interactions, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Groningen, The Netherlands
| | - Paulo Cesar Telles de Souza
- Molecular Microbiology and Structural Biochemistry (MMSB - UMR 5086), CNRS & University of Lyon, Lyon, France
| | - Markus Schwalbe
- Host-Microbe Metabolic Interactions, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Groningen, The Netherlands
| | | | - Warner Hoornenborg
- Department of Behavioral Neurosciences, Cluster Neurobiology, Groningen Institute of for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, The Netherlands
| | - Sieger A. Nelemans
- Department of Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, The Netherlands
| | - Siewert J. Marrink
- Molecular Dynamics, Groningen Biomolecular Sciences and Biotechnology Institute and Zernike Institute for Advanced Materials, University of Groningen, Groningen, The Netherlands
| | - Sahar El Aidy
- Host-Microbe Metabolic Interactions, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Groningen, The Netherlands,CONTACT Sahar El Aidy Host-Microbe Metabolic Interactions, Groningen Biomolecular Sciences and Biotechnology Institute (GBB), University of Groningen, Groningen, The Netherlands
| |
Collapse
|