151
|
Zheng C, Nguyen KK, Vishnivetskiy SA, Gurevich VV, Gurevich EV. Arrestin-3 binds parkin and enhances parkin-dependent mitophagy. J Neurochem 2025; 169:e16043. [PMID: 38196269 PMCID: PMC11231064 DOI: 10.1111/jnc.16043] [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: 05/08/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024]
Abstract
Arrestins were discovered for their role in homologous desensitization of G-protein-coupled receptors (GPCRs). Later non-visual arrestins were shown to regulate several signaling pathways. Some of these pathways require arrestin binding to GPCRs, the regulation of others is receptor independent. Here, we demonstrate that arrestin-3 binds the E3 ubiquitin ligase parkin via multiple sites, preferentially interacting with its RING0 domain. Identification of the parkin domains involved suggests that arrestin-3 likely relieves parkin autoinhibition and/or stabilizes the enzymatically active "open" conformation of parkin. Arrestin-3 binding enhances ubiquitination by parkin of the mitochondrial protein mitofusin-1 and facilitates parkin-mediated mitophagy in HeLa cells. Furthermore, arrestin-3 and its mutant with enhanced parkin binding rescue mitofusin-1 ubiquitination and mitophagy in the presence of the Parkinson's disease-associated R275W parkin mutant, which is defective in both functions. Thus, modulation of parkin activity via arrestin-3 might be a novel strategy of anti-parkinsonian therapy.
Collapse
Affiliation(s)
- Chen Zheng
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | - Kevin K. Nguyen
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, USA
| | | | | | | |
Collapse
|
152
|
Cuello AC, Do Carmo S. The dependence of basal forebrain cholinergic neurons on NGF: The case in Alzheimer pathology. HANDBOOK OF CLINICAL NEUROLOGY 2025; 211:95-122. [PMID: 40340070 DOI: 10.1016/b978-0-443-19088-9.00010-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
This chapter discusses the dependency of basal forebrain cholinergic neurons (BFCNs) on endogenous nerve growth factor (NGF) for the structural and physiologic maintenance of the neuronal cell somata, axonal projections, and terminal synapses. It covers the discovery of NGF and the occurrence of a CNS neurotrophin family and their cognate receptors and their signaling mechanisms. It concludes with a description of the NGF metabolic pathway and its dysregulation in Alzheimer disease (AD) and Down syndrome pathology, explaining the progressive atrophy of BFCNs, which starts at preclinical stages and is reflected in body fluid biomarkers.
Collapse
Affiliation(s)
- A Claudio Cuello
- Department of Pharmacology & Therapeutics, Faculty of Medicine, McGill University, Montreal, QC, Canada; Department of Pharmacology, Oxford University, Oxford, United Kingdom.
| | - Sonia Do Carmo
- Department of Pharmacology & Therapeutics, Faculty of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
153
|
Schöneberg T. Modulating vertebrate physiology by genomic fine-tuning of GPCR functions. Physiol Rev 2025; 105:383-439. [PMID: 39052017 DOI: 10.1152/physrev.00017.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/08/2024] [Accepted: 07/20/2024] [Indexed: 07/27/2024] Open
Abstract
G protein-coupled receptors (GPCRs) play a crucial role as membrane receptors, facilitating the communication of eukaryotic species with their environment and regulating cellular and organ interactions. Consequently, GPCRs hold immense potential in contributing to adaptation to ecological niches and responding to environmental shifts. Comparative analyses of vertebrate genomes reveal patterns of GPCR gene loss, expansion, and signatures of selection. Integrating these genomic data with insights from functional analyses of gene variants enables the interpretation of genotype-phenotype correlations. This review underscores the involvement of GPCRs in adaptive processes, presenting numerous examples of how alterations in GPCR functionality influence vertebrate physiology or, conversely, how environmental changes impact GPCR functions. The findings demonstrate that modifications in GPCR function contribute to adapting to aquatic, arid, and nocturnal habitats, influencing camouflage strategies, and specializing in particular dietary preferences. Furthermore, the adaptability of GPCR functions provides an effective mechanism in facilitating past, recent, or ongoing adaptations in animal domestication and human evolution and should be considered in therapeutic strategies and drug development.
Collapse
Affiliation(s)
- Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, University of Leipzig, Leipzig, Germany
- School of Medicine, University of Global Health Equity, Kigali, Rwanda
| |
Collapse
|
154
|
Calebiro D, Miljus T, O'Brien S. Endomembrane GPCR signaling: 15 years on, the quest continues. Trends Biochem Sci 2025; 50:46-60. [PMID: 39532582 DOI: 10.1016/j.tibs.2024.10.006] [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: 07/08/2024] [Revised: 10/13/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
G-protein-coupled receptors (GPCRs) are the largest family of cell receptors. They mediate the effects of a multitude of endogenous and exogenous cues, are deeply involved in human physiology and disease, and are major pharmacological targets. Whereas GPCRs were long thought to signal exclusively at the plasma membrane, research over the past 15 years has revealed that they also signal via classical G-protein-mediated pathways on membranes of intracellular organelles such as endosomes and the Golgi complex. This review provides an overview of recent advances and emerging concepts related to endomembrane GPCR signaling, as well as ongoing research aimed at a better understanding of its mechanisms, physiological relevance, and potential therapeutic applications.
Collapse
Affiliation(s)
- Davide Calebiro
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK.
| | - Tamara Miljus
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| | - Shannon O'Brien
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| |
Collapse
|
155
|
Xie JJ, Li Y, Wu JH, Fang PX, Li S, Zhou X, Chi CF. FMRFamide G protein-coupled receptors (GPCR) in the cuttlefish Sepiella japonica: Identification, characterization and expression profile. Neuropeptides 2025; 109:102491. [PMID: 39626505 DOI: 10.1016/j.npep.2024.102491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/11/2024] [Accepted: 11/21/2024] [Indexed: 01/06/2025]
Abstract
FMRFamide is a ubiquitous neuromodulator in the animal kingdom. Once FMRFamide or similar neuropeptides bind to their G protein-coupled receptors (GPCR), a series of signal transduction events are triggered, thereby mediating various physiological effects. FMRFamide had been reported to be involved in the regulation of sexual maturation in Sepiella japonica. In this research, the full-length cDNA of FMRFamide G protein-coupled receptor of S. japonica (SjFaGPCR) was cloned. The sequence is 1396 bp long and encodes a protein consisting of 418 amino acid residues, lacking a signal peptide at the N-terminal region. The 3D structure of SjFaGPCR was predicted using Todarodes pacificus rhodopsin as a template, and the result indicated the presence of seven transmembrane regions. Multiple sequence alignments and phylogenetic trees indicated that SjFaGPCR is conserved among invertebrates, and shares highly similar sequence characteristics with other cephalopods. In situ hybridization (ISH) results revealed that significant signals of SjFaGPCR were detected in the central medulla and the granular layer cells of the optic lobe, and were also observed in the supraesophageal and subesophageal masses of the brain. Meanwhile, quantitative real-time PCR (qRT-PCR) results showed that a higher expression level of SjFaGPCR mRNA was detected in the brain and optic lobe of female cuttlefish at stage III and stage VI, and also in the brain (stage V) and optic lobe (stages IV and V) of male cuttlefish than that in other tissues. The co-localization results demonstrated that fluorescence signals of SjFMRFamide and SjFaGPCR were overlapped in HEK293 cells, suggesting a possible interaction between the SjFMRFamide and SjFaGPCR. These findings provide molecular support for further exploring the roles of FMRFamide and FaGPCR in the reproductive regulation of S. japonica.
Collapse
Affiliation(s)
- Jian-Jun Xie
- National and Provincial Joint Engineering Research Centre for Marine Germplasm Resources Exploration and Utilization, School of Marine Science and Technology, Zhejiang Ocean University, 1st Haidanan Road, Changzhi Island, Lincheng, Zhoushan 316022, China
| | - Ying Li
- National and Provincial Joint Engineering Research Centre for Marine Germplasm Resources Exploration and Utilization, School of Marine Science and Technology, Zhejiang Ocean University, 1st Haidanan Road, Changzhi Island, Lincheng, Zhoushan 316022, China
| | - Jun-Hong Wu
- National and Provincial Joint Engineering Research Centre for Marine Germplasm Resources Exploration and Utilization, School of Marine Science and Technology, Zhejiang Ocean University, 1st Haidanan Road, Changzhi Island, Lincheng, Zhoushan 316022, China
| | - Pei-Xuan Fang
- National and Provincial Joint Engineering Research Centre for Marine Germplasm Resources Exploration and Utilization, School of Marine Science and Technology, Zhejiang Ocean University, 1st Haidanan Road, Changzhi Island, Lincheng, Zhoushan 316022, China
| | - Shuang Li
- National and Provincial Joint Engineering Research Centre for Marine Germplasm Resources Exploration and Utilization, School of Marine Science and Technology, Zhejiang Ocean University, 1st Haidanan Road, Changzhi Island, Lincheng, Zhoushan 316022, China
| | - Xu Zhou
- National and Provincial Joint Engineering Research Centre for Marine Germplasm Resources Exploration and Utilization, School of Marine Science and Technology, Zhejiang Ocean University, 1st Haidanan Road, Changzhi Island, Lincheng, Zhoushan 316022, China
| | - Chang-Feng Chi
- National and Provincial Joint Engineering Research Centre for Marine Germplasm Resources Exploration and Utilization, School of Marine Science and Technology, Zhejiang Ocean University, 1st Haidanan Road, Changzhi Island, Lincheng, Zhoushan 316022, China.
| |
Collapse
|
156
|
Wang J, Abrol R, Youkharibache P. Ig or Not Ig? That Is the Question: The Nucleating Supersecondary Structure of the Ig-Fold and the Extended Ig Universe. Methods Mol Biol 2025; 2870:371-396. [PMID: 39543045 DOI: 10.1007/978-1-0716-4213-9_19] [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: 11/17/2024]
Abstract
Observing the omnipresence of the Ig-fold in all domains of life, one may wonder why this fold among all is such a wunderkind of evolution. Culminating in vertebrates, it enables a myriad of functions at the heart of the immune, nervous, vascular, and muscular systems. We suggest the Ig-fold resilience lies in the robust folding of a core supersecondary structure (SSS) that can accommodate a myriad of topological variations. In this chapter, we focus on the core supersecondary structure common to all topostructural variants of the Ig-fold and will see that this pattern can also be found in other β-sandwich folds. It represents a highly resilient central SSS that accommodates a very high plasticity observed among β-sandwiches. We have recently developed a universal numbering system to identify and annotate Ig-domains, Ig-like domains, and what we now call Ig-extended domains, i.e., β-sandwiches that contain and extend the Ig-fold topology (to be published). A universal numbering scheme, common to all topological and structural variants of any domain sharing the Ig-fold, allows a direct comparison of any Ig, Ig-like, and Ig-extended domain in sequence, topology, and structure. This can therefore help understand the robust patterns in Ig-folding and interactions with other Ig or non-Ig proteins, as well as help trace evolutionary patterns of immunoglobulin domains. The universal numbering scheme, called IgStrand, is now at the heart of an algorithm that can label secondary structure elements of the Ig-fold for any topological variant. It is implemented in the open-source web-based iCn3D program from NCBI (Wang, Youkharibache, Zhang, Lanczycki, Geer, Madej, Phan, Ward, Lu, Marchler, Bioinformatics 36:131-135, 2020). Interestingly, that algorithm captures SSS homologies across a very large spectrum of β-sandwiches, and one can envision classifying numerous such sandwiches as "Ig-extended" domains and their variable topological arrangements. In this chapter, we go through examples of Ig, Ig-like, and Ig-extended domains as in a journey through cells: in the cell nucleus, in the cytoplasm, or on extracellular regions of cell surface receptors, and in viruses.
Collapse
Affiliation(s)
- Jiyao Wang
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Ravinder Abrol
- Department of Chemistry and Biochemistry, California State University, Northridge, CA, USA
| | - Philippe Youkharibache
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
157
|
Kajino K, Sugai T, Kise R, Suzuki R, Tokuda A, Sekiya Y, Kakumoto T, Katamoto R, Kutsumura N, Nagumo Y, Inoue A, Saitoh T. Structure-Signal Relationships of the δ-Opioid-Receptor (DOR)-Selective Agonist KNT-127-Part I: Impact of the Morphinan Skeleton on the G-Protein-Biased DOR Agonism. Chem Pharm Bull (Tokyo) 2025; 73:246-256. [PMID: 40159181 DOI: 10.1248/cpb.c25-00012] [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: 04/02/2025]
Abstract
The δ-opioid receptor (DOR) is a promising target for developing novel analgesics due to its lower risk of causing side effects compared to the μ-opioid receptor (MOR), which is commonly associated with dependence, respiratory depression, and other adverse effects. KNT-127, a DOR-selective agonist with a morphinan skeleton, offers analgesic and antidepressant benefits without inducing convulsions at therapeutic doses, unlike the conventional DOR agonist SNC80. While previous studies have suggested that KNT-127 exhibits reduced β-arrestin recruitment, a signaling pathway implicated in adverse opioid effects, the ligand structural basis for this biased signaling remains unclear. In this study, we explored the structure-signal relationships of KNT-127, focusing on its quinoline moiety, which is known to serve as an address domain responsible for DOR selectivity. Modifying the quinoline moiety by removing the aromatic rings reduced DOR selectivity and potency in relation to G-protein activation while diminishing both the potency and efficacy of β-arrestin recruitment. These results suggest that the morphinan skeleton is critical for reduced β-arrestin recruitment, while the quinoline moiety differentially modulates G-protein activation and β-arrestin recruitment. Together, our study expands the message-address concept, previously limited to receptor selectivity, by providing structural insights into the G-protein-biased agonism of DOR agonists, thereby guiding the design of safer DOR-targeting therapeutics.
Collapse
Affiliation(s)
- Keita Kajino
- Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Tomoya Sugai
- Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Ryoji Kise
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Riko Suzuki
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
| | - Akihisa Tokuda
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Yuki Sekiya
- Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Tomoya Kakumoto
- Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Risako Katamoto
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Noriki Kutsumura
- Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Yasuyuki Nagumo
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan
- Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimo-Adachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Tsuyoshi Saitoh
- Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8571, Japan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
- Division of Biomedical Science, Institute of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
158
|
Caniceiro AB, Orzeł U, Rosário-Ferreira N, Filipek S, Moreira IS. Leveraging Artificial Intelligence in GPCR Activation Studies: Computational Prediction Methods as Key Drivers of Knowledge. Methods Mol Biol 2025; 2870:183-220. [PMID: 39543036 DOI: 10.1007/978-1-0716-4213-9_10] [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: 11/17/2024]
Abstract
G protein-coupled receptors (GPCRs) are key molecules involved in cellular signaling and are attractive targets for pharmacological intervention. This chapter is designed to explore the range of algorithms used to predict GPCRs' activation states, while also examining the pharmaceutical implications of these predictions. Our primary objective is to show how artificial intelligence (AI) is key in GPCR research to reveal the intricate dynamics of activation and inactivation processes, shedding light on the complex regulatory mechanisms of this vital protein family. We describe several computational strategies that leverage diverse structural data from the Protein Data Bank, molecular dynamic simulations, or ligand-based methods to predict the activation states of GPCRs. We demonstrate how the integration of AI into GPCR research not only enhances our understanding of their dynamic properties but also presents immense potential for driving pharmaceutical research and development, offering promising new avenues in the search for newer, better therapeutic agents.
Collapse
Affiliation(s)
- Ana B Caniceiro
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Urszula Orzeł
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Nícia Rosário-Ferreira
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Sławomir Filipek
- Faculty of Chemistry, Biological and Chemical Research Centre, University of Warsaw, Warsaw, Poland
| | - Irina S Moreira
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
- CNC-UC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
159
|
Cheng D, Guo Y, Lyu J, Liu Y, Xu W, Zheng W, Wang Y, Qiao P. Advances and challenges in preparing membrane proteins for native mass spectrometry. Biotechnol Adv 2025; 78:108483. [PMID: 39571766 DOI: 10.1016/j.biotechadv.2024.108483] [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: 05/31/2024] [Revised: 10/07/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
Native mass spectrometry (nMS) is becoming a crucial tool for analyzing membrane proteins (MPs), yet challenges remain in solubilizing and stabilizing their native conformations while resolving and characterizing the heterogeneity introduced by post-translational modifications and ligand binding. This review highlights recent advancements and persistent challenges in preparing MPs for nMS. Optimizing detergents and additives can significantly reduce sample heterogeneity and surface charge, enhancing MP signal quality and structural preservation in nMS. A strategic workflow incorporating affinity capture, stabilization agents, and size-exclusion chromatography to remove unfolded species demonstrates success in improving nMS characterization. Continued development of customized detergents and reagents tailored for specific MPs may further minimize heterogeneity and boost signals. Instrumental advances are also needed to elucidate more dynamically complex and labile MPs. Effective sample preparation workflows may provide insights into MP structures, dynamics, and interactions underpinning membrane biology. With ongoing methodological innovation, nMS shows promise to complement biophysical studies and facilitate drug discovery targeting this clinically important yet technically demanding protein class.
Collapse
Affiliation(s)
- Di Cheng
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yi Guo
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Jixing Lyu
- Department of Chemistry, Texas A&M University, College Station, TX 77843, USA
| | - Yang Liu
- Regenxbox In., Rockville, MD 20850, USA
| | - Wenhao Xu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Weiyi Zheng
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yuchen Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Pei Qiao
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China.
| |
Collapse
|
160
|
Pfersdorf F, Romanazzi L, Rosenkilde MM, Gustavsson M. Regulation of the chemokine receptors CXCR4 and ACKR3 by receptor activity-modifying proteins. J Biol Chem 2025; 301:108055. [PMID: 39662834 PMCID: PMC11760809 DOI: 10.1016/j.jbc.2024.108055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 11/06/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024] Open
Abstract
The chemokine CXCL12 and its two cognate receptors-CXCR4 and ACKR3-are key players in various homeostatic and pathophysiological processes, including embryonic development, autoimmune diseases, tissue repair, and cancer. Recent reports identified an interaction of CXCR4 and ACKR3 with receptor activity-modifying proteins (RAMPs), and RAMP3 has been shown to facilitate ACKR3's recycling properties. Yet, the functional effects of RAMPs on the CXCL12 signaling axis remain largely elusive. Here, we characterize the effects of RAMPs on CXCR4 and ACKR3 function. We show that, in the absence of a ligand, RAMPs do not affect the cell membrane localization or constitutive internalization of the two receptors. RAMP3 inhibits ligand-stimulated internalization of ACKR3, which retains the receptor at the membrane and inhibits its ability to scavenge CXCL12. In addition, while cAMP inhibition by CXCR4 is unaffected by RAMPs, basal and ligand-stimulated β-arrestin recruitment to both CXCR4 and ACKR3 is reduced in the presence of RAMP3 due to complex formation at the cell surface. The effects on ACKR3 are observed for chemokine, small molecule, and peptide agonists as well as for a N-terminal truncated receptor variant, suggesting that RAMP regulation involves contacts with the transmembrane domain of the receptor. Taken together, our results show that RAMPs regulate the CXCL12 signaling axis by directly interfering with receptor function. These findings could have direct implications for the interplay between receptors in vivo as well as future drug design in the therapeutic targeting of the CXCL12 signaling axis.
Collapse
Affiliation(s)
- Fabian Pfersdorf
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lucas Romanazzi
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Martin Gustavsson
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
161
|
Sun Y, Hasbi A, George SR. G Protein-Coupled Receptor Heteromers in Brain: Functional and Therapeutic Importance in Neuropsychiatric Disorders. Annu Rev Pharmacol Toxicol 2025; 65:215-236. [PMID: 39847466 DOI: 10.1146/annurev-pharmtox-061724-080727] [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: 01/25/2025]
Abstract
G protein-coupled receptors (GPCRs) represent the largest family of plasma membrane proteins targeted for therapeutic development. For decades, GPCRs were investigated as monomeric entities during analysis of their pharmacology or signaling and during drug development. However, a considerable body of evidence now indicates that GPCRs function as dimers or higher-order oligomers. Greater acceptance of oligomerization occurred with the recognition that GPCR interactions form heteromeric receptor complexes, which was validated in vivo, often with pharmacologic, signaling, and functional properties distinct from the constituent protomers. GPCR heteromerization is reviewed in the context of brain disorders, with examples illustrating their functional implication in diverse neuropsychiatric and neurodegenerative disorders, making them an enormous unexploited resource for selective pharmacotherapy target identification. The strategies for development of heteromer-selective ligands are discussed as a new opportunity to precisely target the function of a receptor complex with greater specificity, in contrast to the classical ligands targeting individual receptors.
Collapse
Affiliation(s)
- Yalin Sun
- Department of Pharmacology and Toxicology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada;
| | - Ahmed Hasbi
- Department of Pharmacology and Toxicology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada;
| | - Susan R George
- Department of Pharmacology and Toxicology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada;
- Department of Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
162
|
Pearce A, Redfern-Nichols T, Wills E, Rosa M, Manulak I, Sisk C, Huang X, Atakpa-Adaji P, Prole DL, Ladds G. Quantitative approaches for studying G protein-coupled receptor signalling and pharmacology. J Cell Sci 2025; 138:JCS263434. [PMID: 39810711 PMCID: PMC11828474 DOI: 10.1242/jcs.263434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025] Open
Abstract
G protein-coupled receptor (GPCR) signalling pathways underlie numerous physiological processes, are implicated in many diseases and are major targets for therapeutics. There are more than 800 GPCRs, which together transduce a vast array of extracellular stimuli into a variety of intracellular signals via heterotrimeric G protein activation and multiple downstream effectors. A key challenge in cell biology research and the pharmaceutical industry is developing tools that enable the quantitative investigation of GPCR signalling pathways to gain mechanistic insights into the varied cellular functions and pharmacology of GPCRs. Recent progress in this area has been rapid and extensive. In this Review, we provide a critical overview of these new, state-of-the-art approaches to investigate GPCR signalling pathways. These include novel sensors, Förster or bioluminescence resonance energy transfer assays, libraries of tagged G proteins and transcriptional reporters. These approaches enable improved quantitative studies of different stages of GPCR signalling, including GPCR activation, G protein activation, second messenger (cAMP and Ca2+) signalling, β-arrestin recruitment and the internalisation and intracellular trafficking of GPCRs.
Collapse
Affiliation(s)
- Abigail Pearce
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Theo Redfern-Nichols
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Edward Wills
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Matthew Rosa
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Iga Manulak
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Claudia Sisk
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Xianglin Huang
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Peace Atakpa-Adaji
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - David L. Prole
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| |
Collapse
|
163
|
Guo Q, Huo Y, Liu Q, Zhou S, Xiao Y. Ruxolitinib as a CaMKII inhibitor for treatment of cardiac arrhythmias: Applications and prospects. Heart Rhythm 2025; 22:231-239. [PMID: 39111609 DOI: 10.1016/j.hrthm.2024.07.118] [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: 05/05/2024] [Revised: 07/20/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024]
Abstract
Recent studies have highlighted the critical role of calcium/calmodulin-dependent protein kinase II (CaMKII) overactivation in the pathogenesis of various cardiac arrhythmias. Ruxolitinib, a Janus kinase inhibitor widely used for the treatment of myelofibrosis and acute graft-vs-host disease, has expanded its research horizons to include its potential as a CaMKII inhibitor in the treatment of cardiac arrhythmias. This article reviews the basic pharmacologic properties of ruxolitinib and delves into the role of CaMKII in cardiac arrhythmias, including its structural fundamentals, activation mechanisms, and association with arrhythmic conditions. Furthermore, the current state of CaMKII inhibitor research is discussed, with a special focus on the advances and clinical potential of ruxolitinib in this field. Studies indicate that ruxolitinib effectively inhibits CaMKII activity and has therapeutic potential against cardiac arrhythmias in animal models and at the cellular level. In addition, we address the critical issues that need to be resolved before the clinical application of ruxolitinib in arrhythmia treatment, including dosage concerns, long-term inhibitory effects, potential impacts on the nervous system, and efficacy across different types of arrhythmias. Future research directions involve further exploration of the clinical application potential of ruxolitinib, particularly in diseases such as heart failure, hypertrophic cardiomyopathy, dilated cardiomyopathy, and ischemic arrhythmias. In summary, the efficacy, low toxicity, and safety profile of ruxolitinib as a CaMKII inhibitor in the treatment of cardiac arrhythmias suggest a promising future for its development as a therapeutic drug in this domain.
Collapse
Affiliation(s)
- Qingbo Guo
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha, Hunan, PR China; Xiangya School of Medicine, Central South University, Changsha, Hunan, PR China
| | - Yiran Huo
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha, Hunan, PR China; Xiangya School of Medicine, Central South University, Changsha, Hunan, PR China
| | - Qiming Liu
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Shenghua Zhou
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha, Hunan, PR China
| | - Yichao Xiao
- Department of Cardiovascular Medicine, Second Xiangya Hospital of Central South University, Changsha, Hunan, PR China.
| |
Collapse
|
164
|
Salim E, Hori A, Matsubara K, Takano-Shimizu T, Pratomo AR, Marianne M, Syahputra A, Husori DI, Inoue A, Abdullah MA, Shamsudin NF, Rullah K, Kuraishi T. Detection of Human GPCR Activity in Drosophila S2 Cells Using the Tango System. Int J Mol Sci 2024; 26:202. [PMID: 39796060 PMCID: PMC11720185 DOI: 10.3390/ijms26010202] [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: 12/03/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
G protein-coupled receptors (GPCRs) are essential cell surface proteins involved in transducing extracellular signals into intracellular responses, regulating various physiological processes. This study validated the use of the Tango assay, a sensitive method for detecting GPCR activation, in Drosophila Schneider 2 (S2) cells, focusing on the human Dopamine Receptor D4 (DRD4). Plasmids encoding the LexA-tagged human DRD4 receptor and a luciferase reporter were co-transfected into Drosophila S2 cells and stimulated with dopamine. Receptor activation was measured by quantifying the luciferase activity. The system showed high specificity for dopamine, with no activation in response to octopamine, a non-ligand for DRD4. Furthermore, the system effectively detects activation by a novel compound. These results demonstrate that Drosophila S2 cells, coupled with the Tango assay, provide a viable model for studying human GPCR function and ligand specificity. This system enables the rapid screening of potential GPCR ligands in a cost-effective cellular model.
Collapse
Affiliation(s)
- Emil Salim
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Osaka 920-1192, Japan; (A.H.); (A.R.P.)
- Department of Pharmacology and Clinical/Community Pharmacy, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia; (M.M.); (D.I.H.)
| | - Aki Hori
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Osaka 920-1192, Japan; (A.H.); (A.R.P.)
| | - Kohei Matsubara
- KYOTO Drosophila Stock Center, Kyoto Institute of Technology, Saga Ippongi-cho 1, Ukyo-ku, Kyoto 616-8354, Japan; (K.M.); (T.T.-S.)
| | - Toshiyuki Takano-Shimizu
- KYOTO Drosophila Stock Center, Kyoto Institute of Technology, Saga Ippongi-cho 1, Ukyo-ku, Kyoto 616-8354, Japan; (K.M.); (T.T.-S.)
| | - Andre Rizky Pratomo
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Osaka 920-1192, Japan; (A.H.); (A.R.P.)
| | - Marianne Marianne
- Department of Pharmacology and Clinical/Community Pharmacy, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia; (M.M.); (D.I.H.)
| | - Armia Syahputra
- Departement of Periodontology, Faculty of Dentistry, Universitas Sumatera Utara, Medan 20155, Indonesia;
| | - Dadang Irfan Husori
- Department of Pharmacology and Clinical/Community Pharmacy, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia; (M.M.); (D.I.H.)
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3, Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan;
- Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimo-Adachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Maryam Aisyah Abdullah
- Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, Bandar Indera Mahkota, Kuantan 25200, Pahang, Malaysia; (M.A.A.); (N.F.S.); (K.R.)
| | - Nur Farisya Shamsudin
- Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, Bandar Indera Mahkota, Kuantan 25200, Pahang, Malaysia; (M.A.A.); (N.F.S.); (K.R.)
| | - Kamal Rullah
- Department of Pharmaceutical Chemistry, Kulliyyah of Pharmacy, International Islamic University Malaysia, Bandar Indera Mahkota, Kuantan 25200, Pahang, Malaysia; (M.A.A.); (N.F.S.); (K.R.)
| | - Takayuki Kuraishi
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Osaka 920-1192, Japan; (A.H.); (A.R.P.)
| |
Collapse
|
165
|
Niebrügge N, Trovato O, Praschberger R, Lieb A. Disease-Associated Dopamine Receptor D2 Variants Exhibit Functional Consequences Depending on Different Heterotrimeric G-Protein Subunit Combinations. Biomedicines 2024; 13:46. [PMID: 39857630 PMCID: PMC11761627 DOI: 10.3390/biomedicines13010046] [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: 12/05/2024] [Revised: 12/20/2024] [Accepted: 12/26/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Dopamine receptors (DRs) are G-protein-coupled receptors (GPCRs) found in the central nervous system (CNS). DRs are essential for mediating various downstream signaling cascades and play a critical role in regulating the dopaminergic nigrostriatal pathway, which is involved in motor control. Recently, mutations in DRD2 (WT), p.Ile212Phe (I212F), and p.Met345Arg (M345R) have been associated with hyperkinetic movement disorders and shown to alter heterotrimeric G-protein complex signaling and β-arrestin recruitment. Methods: To conduct a detailed investigation of the I212F and M345R functional phenotypes, we used the TRansdUcer PATHway (TRUPATH) assay to study heterotrimeric G-protein recruitment and the Parallel Receptorome Expression and Screening via Transcriptional Output (PRESTO-Tango) assay to evaluate transcriptional activation following arrestin translocation for β-arrestin recruitment. Results: In our study, we could confirm the reported mutant's loss-of-function phenotype in β-arrestin 2 recruitment (reduced agonist potency and decreased maximal signaling efficacy in comparison to the WT). However, a detailed analysis of basal/constitutive activity also revealed a gain-of-function phenotype for mutant M345R. For a more comprehensive investigation of heterotrimeric G-protein complex signaling, we investigated the impact of WT mutants in combination with (i) a specifically suggested assay, and (ii) the most abundantly expressed heterotrimeric G-protein complex combinations in WT receptor-enriched regions. We were able to confirm the reported gain-of-function phenotype by Rodriguez-Contreras et al. and extend it by the use of the most abundant heterotrimeric G-protein subunits, GαoA and Gαi1, β1 and β2, and γ3 and γ7, in mouse and human basal ganglia. Conclusions: Although our results indicate that the interaction of the two variants with the most highly expressed heterotrimeric G-protein complex subunit combinations also results in a gain-of-function phenotype, they also clearly demonstrate that the phenotype can be significantly altered, dependent on heterotrimeric G-protein complex expression.
Collapse
Affiliation(s)
- Nele Niebrügge
- Institute of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Olga Trovato
- Institute of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Roman Praschberger
- Institute of Human Genetic, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Andreas Lieb
- Institute of Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
166
|
Vinogradova TM, Lakatta EG. Ca 2+/Calmodulin-Dependent Protein Kinase II (CaMKII) Regulates Basal Cardiac Pacemaker Function: Pros and Cons. Cells 2024; 14:3. [PMID: 39791704 PMCID: PMC11719954 DOI: 10.3390/cells14010003] [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: 11/16/2024] [Revised: 12/14/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025] Open
Abstract
The spontaneous firing of the sinoatrial (SA) node, the physiological pacemaker of the heart, is generated within sinoatrial nodal cells (SANCs) and is regulated by a "coupled-clock" pacemaker system, which integrates a "membrane clock", the ensemble of ion channel currents, and an intracellular "Ca2+ clock", sarcoplasmic reticulum-generated local submembrane Ca2+ releases via ryanodine receptors. The interactions within a "coupled-clock" system are modulated by phosphorylation of surface membrane and sarcoplasmic reticulum proteins. Though the essential role of a high basal cAMP level and PKA-dependent phosphorylation for basal spontaneous SANC firing is well recognized, the role of basal CaMKII-dependent phosphorylation remains uncertain. This is a critical issue with respect to how cardiac pacemaker cells fire spontaneous action potentials. This review aspires to explain and unite apparently contradictory results of pharmacological studies in the literature that have demonstrated a fundamental role of basal CaMKII activation for basal cardiac pacemaker function, as well as studies in mice with genetic CaMKII inhibition which have been interpreted to indicate that basal spontaneous SANC firing is independent of CaMKII activation. The assessment of supporting and opposing data regarding CaMKII effects on phosphorylation of Ca2+-cycling proteins and spontaneous firing of SANC in the basal state leads to the necessary conclusion that CaMKII activity and CaMKII-dependent phosphorylation do regulate basal cardiac pacemaker function.
Collapse
Affiliation(s)
- Tatiana M. Vinogradova
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institute of Health, Baltimore, MD 21224, USA;
| | | |
Collapse
|
167
|
Zhang P, Deng H, Lan X, Shen P, Bai Z, Huangfu C, Wang N, Xiao C, Gao Y, Sun Y, Li J, Guo J, Zhou W, Gao Y. Tetramethylpyrazine Protects Against Chronic Hypobaric Hypoxia-Induced Cardiac Dysfunction by Inhibiting CaMKII Activation in a Mouse Model Study. Int J Mol Sci 2024; 26:54. [PMID: 39795913 PMCID: PMC11720575 DOI: 10.3390/ijms26010054] [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: 11/25/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
Chronic exposure to high altitudes causes pathophysiological cardiac changes that are characterized by cardiac dysfunction, cardiac hypertrophy, and decreased energy reserves. However, finding specific pharmacological interventions for these pathophysiological changes is challenging. In this study, we identified tetramethylpyrazine (TMP) as a promising drug candidate for cardiac dysfunction caused by simulated high-altitude exposure. By utilizing hypobaric chambers to simulate high-altitude environments, we found that TMP improved cardiac function, alleviated cardiac hypertrophy, and reduced myocardial injury in hypobaric hypoxic mice. RNA sequencing showed that TMP also upregulated heart-contraction-related genes that were suppressed by hypobaric hypoxia exposure. Mechanistically, TMP inhibited hypobaric hypoxia-induced cardiac Ca2+/calmodulin-dependent kinase II (CaMKII) activation and exerted cardioprotective effects by inhibiting CaMKII. Our data suggest that TMP application may be a promising approach for treating high-altitude-induced cardiac dysfunction, and they highlight the crucial role of CaMKII in hypobaric hypoxia-induced cardiac pathophysiology.
Collapse
Affiliation(s)
- Pengfei Zhang
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (P.Z.); (H.D.); (X.L.); (P.S.); (Z.B.); (C.H.); (N.W.); (C.X.); (Y.G.); (Y.S.); (J.L.); (J.G.)
| | - Huifang Deng
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (P.Z.); (H.D.); (X.L.); (P.S.); (Z.B.); (C.H.); (N.W.); (C.X.); (Y.G.); (Y.S.); (J.L.); (J.G.)
| | - Xiong Lan
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (P.Z.); (H.D.); (X.L.); (P.S.); (Z.B.); (C.H.); (N.W.); (C.X.); (Y.G.); (Y.S.); (J.L.); (J.G.)
| | - Pan Shen
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (P.Z.); (H.D.); (X.L.); (P.S.); (Z.B.); (C.H.); (N.W.); (C.X.); (Y.G.); (Y.S.); (J.L.); (J.G.)
| | - Zhijie Bai
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (P.Z.); (H.D.); (X.L.); (P.S.); (Z.B.); (C.H.); (N.W.); (C.X.); (Y.G.); (Y.S.); (J.L.); (J.G.)
| | - Chaoji Huangfu
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (P.Z.); (H.D.); (X.L.); (P.S.); (Z.B.); (C.H.); (N.W.); (C.X.); (Y.G.); (Y.S.); (J.L.); (J.G.)
| | - Ningning Wang
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (P.Z.); (H.D.); (X.L.); (P.S.); (Z.B.); (C.H.); (N.W.); (C.X.); (Y.G.); (Y.S.); (J.L.); (J.G.)
| | - Chengrong Xiao
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (P.Z.); (H.D.); (X.L.); (P.S.); (Z.B.); (C.H.); (N.W.); (C.X.); (Y.G.); (Y.S.); (J.L.); (J.G.)
| | - Yehui Gao
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (P.Z.); (H.D.); (X.L.); (P.S.); (Z.B.); (C.H.); (N.W.); (C.X.); (Y.G.); (Y.S.); (J.L.); (J.G.)
| | - Yue Sun
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (P.Z.); (H.D.); (X.L.); (P.S.); (Z.B.); (C.H.); (N.W.); (C.X.); (Y.G.); (Y.S.); (J.L.); (J.G.)
| | - Jiamiao Li
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (P.Z.); (H.D.); (X.L.); (P.S.); (Z.B.); (C.H.); (N.W.); (C.X.); (Y.G.); (Y.S.); (J.L.); (J.G.)
| | - Jie Guo
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (P.Z.); (H.D.); (X.L.); (P.S.); (Z.B.); (C.H.); (N.W.); (C.X.); (Y.G.); (Y.S.); (J.L.); (J.G.)
| | - Wei Zhou
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (P.Z.); (H.D.); (X.L.); (P.S.); (Z.B.); (C.H.); (N.W.); (C.X.); (Y.G.); (Y.S.); (J.L.); (J.G.)
| | - Yue Gao
- Beijing Institute of Radiation Medicine, Beijing 100850, China; (P.Z.); (H.D.); (X.L.); (P.S.); (Z.B.); (C.H.); (N.W.); (C.X.); (Y.G.); (Y.S.); (J.L.); (J.G.)
- State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
168
|
Petrovic I, Grzesiek S, Isaikina P. Advances in the molecular understanding of GPCR-arrestin complexes. Biochem Soc Trans 2024; 52:2333-2342. [PMID: 39508463 DOI: 10.1042/bst20240170] [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: 07/16/2024] [Revised: 09/17/2024] [Accepted: 10/07/2024] [Indexed: 11/15/2024]
Abstract
Arrestins are essential proteins for the regulation of G protein-coupled receptors (GPCRs). They mediate GPCR desensitization after the activated receptor has been phosphorylated by G protein receptor kinases (GRKs). In addition, GPCR-arrestin interactions may trigger signaling pathways that are distinct and independent from G proteins. The non-visual GPCRs encompass hundreds of receptors with varying phosphorylation patterns and amino acid sequences, which are regulated by only two human non-visual arrestin isoforms. This review describes recent findings on GPCR-arrestin complexes, obtained by structural techniques, biophysical, biochemical, and cellular assays. The solved structures of complete GPCR-arrestin complexes are of limited resolution ranging from 3.2 to 4.7 Å and reveal a high variability in the relative receptor-arrestin orientation. In contrast, biophysical and functional data indicate that arrestin recruitment, activation and GPCR-arrestin complex stability depend on the receptor phosphosite sequence patterns and density. At present, there is still a manifest lack of high-resolution structural and dynamical information on the interactions of native GPCRs with both GRKs and arrestins, which could provide a detailed molecular understanding of the genesis of receptor phosphorylation patterns and the specificity GPCR-arrestin interactions. Such insights seem crucial for progress in the rational design of advanced, arrestin-specific therapeutics.
Collapse
Affiliation(s)
- Ivana Petrovic
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | | | - Polina Isaikina
- Center for Life Sciences, Paul Scherrer Institut, CH-5232 Villigen, Switzerland
| |
Collapse
|
169
|
Chai Z, Silverman D, Li S, Bina P, Yau KW. Dark continuous noise from visual pigment as a major mechanism underlying rod-cone difference in light sensitivity. Proc Natl Acad Sci U S A 2024; 121:e2418031121. [PMID: 39656211 PMCID: PMC11665912 DOI: 10.1073/pnas.2418031121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/07/2024] [Indexed: 01/15/2025] Open
Abstract
Retinal rods and cones underlie scotopic and photopic vision, respectively. Their pigments exhibit spontaneous isomerizations (quantal noise) in darkness due to intrinsic thermal energy. This quantal noise, albeit exceedingly low in rods, dictates the light threshold for scotopic vision. The same quantal noise in cones, however, is too low to explain the much higher diurnal light threshold. Separately, a dark continuous noise is present in rods, long accepted to originate from an intrinsic random activation of the cyclic guanosine monophosphate (cGMP)-phosphodiesterase enzyme mediating phototransduction downstream of the pigment. Here, we report the surprising finding that most of this rod dark continuous noise actually originates from rhodopsin itself. Importantly, we found the same continuous noise with a much higher magnitude from cone pigments. The rod and cone continuous noises are apparently both associated with a hitherto unrecognized "metastable" pigment conformational state physiologically resembling that in apo-opsin (opsin devoid of chromophore) and is intermittently active for very brief moments. The cone holopigment's high continuous noise is expected to act as an intrinsic equivalent light and adapt the cone dramatically, accounting for a major part of the light-sensitivity difference between rods and cones in darkness.
Collapse
Affiliation(s)
- Zuying Chai
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Daniel Silverman
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Sihan Li
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Parinaz Bina
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - King-Wai Yau
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD21205
| |
Collapse
|
170
|
Zheng Z, Ke L, Ye S, Shi P, Yao H. Pharmacological Mechanisms of Cryptotanshinone: Recent Advances in Cardiovascular, Cancer, and Neurological Disease Applications. Drug Des Devel Ther 2024; 18:6031-6060. [PMID: 39703195 PMCID: PMC11658958 DOI: 10.2147/dddt.s494555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024] Open
Abstract
Cryptotanshinone (CTS) is an important active ingredient of Salvia miltiorrhiza Bge. In recent years, its remarkable pharmacological effects have triggered extensive and in-depth studies. The aim of this study is to retrieve the latest research progress on CTS and provide prospects for future research. The selection of literature for inclusion, data extraction and methodological quality assessment were discussed. Studies included (1) physicochemical and ADME/Tox properties, (2) pharmacological effects and mechanism, (3) conclusion and bioinformatics analysis. A total of 915 titles and abstracts were screened, resulting in 184 papers used in this review; CTS has shown therapeutic effects on a variety of diseases by modulating multiple molecular pathways. For example, CTS primarily targets NF-κB pathway and MAPK pathway to have a therapeutic role in cardiovascular diseases; in cancer, CTS shows superior efficacy through the PI3K/Akt/mTOR pathway and the JAK/STAT pathway; CTS act on the Nrf2/HO-1 pathway to combat neurological diseases. In addition, key targets of CTS were predicted by bioinformatics analysis, referring to disease ontology (DO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene ontology (GO) enrichment analysis, with R Studio; AKT1, MAPK1, STAT3, P53 and EGFR are predicted to be the key targets of CTS against diseases. The key proteins were then docked by Autodock software to preliminarily assess their binding activities. This review provided new insights into research of CTS and its potential applications in the future, and especially the targets and directly binding modes for CTS are waiting to be investigated.
Collapse
Affiliation(s)
- Ziyao Zheng
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Liyuan Ke
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Shumin Ye
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| | - Peiying Shi
- Department of Traditional Chinese Medicine Resource and Bee Products, College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, 350002, People’s Republic of China
| | - Hong Yao
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, Fujian Medical University, Fuzhou, 350122, People’s Republic of China
| |
Collapse
|
171
|
Seyedabadi M, Gurevich VV. Flavors of GPCR signaling bias. Neuropharmacology 2024; 261:110167. [PMID: 39306191 DOI: 10.1016/j.neuropharm.2024.110167] [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: 04/23/2024] [Revised: 08/06/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024]
Abstract
GPCRs are inherently flexible molecules existing in an equilibrium of multiple conformations. Binding of GPCR agonists shifts this equilibrium. Certain agonists can increase the fraction of active-like conformations that predispose the receptor to coupling to a particular signal transducer or a select group of transducers. Such agonists are called biased, in contrast to balanced agonists that facilitate signaling via all transducers the receptor couples to. These biased agonists preferentially channel the signaling of a GPCR to particular G proteins, GRKs, or arrestins. Preferential activation of particular G protein or arrestin subtypes can be beneficial, as it would reduce unwanted on-target side effects, widening the therapeutic window. However, biasing GPCRs has two important limitations: a) complete bias is impossible due to inherent flexibility of GPCRs; b) receptor-independent functions of signal transducer proteins cannot be directly affected by GPCR ligands or differential receptor barcoding by GRK phosphorylation. This article is part of the Special Issue on "Ligand Bias".
Collapse
Affiliation(s)
- Mohammad Seyedabadi
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Ave South, PRB, Rm. 417D, Nashville, TN, 37232, USA.
| |
Collapse
|
172
|
Kurt H, Akyol A, Son CD, Zheng C, Gado I, Meli M, Ferrandi EE, Bassanini I, Vasile F, Gurevich VV, Nebol A, Cagavi E, Morra G, Sensoy O. A small molecule enhances arrestin-3 binding to the β 2-adrenergic receptor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.12.628161. [PMID: 39713392 PMCID: PMC11661165 DOI: 10.1101/2024.12.12.628161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
G protein-coupled receptor (GPCR) signaling is terminated by arrestin binding to a phosphorylated receptor. Binding propensity has been shown to be modulated by stabilizing the pre-activated state of arrestin through point mutations or C-tail truncation. Here, we hypothesize that pre-activated rotated states can be stabilized by small molecules, and this can promote binding to phosphorylation-deficient receptors, which underly a variety of human disorders. We performed virtual screening on druggable pockets identified on pre-activated conformations in Molecular Dynamics trajectories of arrestin-3, and found a compound targeting an activation switch, the back loop at the inter-domain interface. According to our model, consistent with available biochemical and structural data, the compound destabilized the ionic lock between the finger and the back loop, and enabled transition of the `gate loop` towards the pre-activated state, which stabilizes pre-activated inter-domain rotation. The predicted binding pocket is consistent with saturation-transfer difference NMR data indicating close contact between the piperazine moiety of the compound and C/finger loops. The compound increases in-cell arrestin-3 binding to phosphorylation-deficient and wild-type β2-adrenergic receptor, but not to muscarinic M2 receptor, as verified by FRET and NanoBiT. This study demonstrates that the back loop can be targeted to modulate interaction of arrestin with phosphorylation-deficient GPCRs in a receptor-specific manner.
Collapse
Affiliation(s)
- Han Kurt
- Istanbul Medipol University, Graduate School of Engineering and Natural Sciences, 34810, Istanbul, Turkey
- present address: University of Cagliari, Department of Physics, Cittadella Universitaria, I-09042 Monserrato (CA), Italy
| | - Ali Akyol
- The Middle East Technical University, Department of Biological Sciences, Ankara 06800, Turkey
| | - Cagdas Devrim Son
- The Middle East Technical University, Department of Biological Sciences, Ankara 06800, Turkey
| | - Chen Zheng
- Vanderbilt University, Department of Pharmacology, 37232, Nashville, TN, USA
| | - Irene Gado
- University of Milano, Department of Chemistry, via Golgi 19, 20131 Milano, Italy
| | - Massimiliano Meli
- Consiglio Nazionale delle Ricerche, Istituto di Scienze e Tecnologie Chimiche “G. Natta” SCITEC, via Mario Bianco 9, 20131, Milano, Italy
| | - Erica Elisa Ferrandi
- Consiglio Nazionale delle Ricerche, Istituto di Scienze e Tecnologie Chimiche “G. Natta” SCITEC, via Mario Bianco 9, 20131, Milano, Italy
| | - Ivan Bassanini
- Consiglio Nazionale delle Ricerche, Istituto di Scienze e Tecnologie Chimiche “G. Natta” SCITEC, via Mario Bianco 9, 20131, Milano, Italy
| | - Francesca Vasile
- University of Milano, Department of Chemistry, via Golgi 19, 20131 Milano, Italy
| | | | - Aylin Nebol
- Istanbul Medipol University, Research Institute for Health Sciences and Technologies (SABITA), Regenerative and Restorative Medical Research Center (REMER), 34810, Istanbul, Turkey
- Istanbul Medipol University, Institute for Health Sciences, Medical Biology and Genetics Program, 34810, Istanbul, Turkey
- Istanbul Medipol University, School of Medicine, Department of Medical Biology, 34810, Istanbul, Turkey
| | - Esra Cagavi
- Istanbul Medipol University, Research Institute for Health Sciences and Technologies (SABITA), Regenerative and Restorative Medical Research Center (REMER), 34810, Istanbul, Turkey
- Istanbul Medipol University, Institute for Health Sciences, Medical Biology and Genetics Program, 34810, Istanbul, Turkey
- Istanbul Medipol University, School of Medicine, Department of Medical Biology, 34810, Istanbul, Turkey
| | - Giulia Morra
- Consiglio Nazionale delle Ricerche, Istituto di Scienze e Tecnologie Chimiche “G. Natta” SCITEC, via Mario Bianco 9, 20131, Milano, Italy
| | - Ozge Sensoy
- Istanbul Medipol University, School of Engineering and Natural Sciences, Department of Biomedical Engineering, 34810, Istanbul, Turkey
- Istanbul Medipol University, Research Institute for Health Sciences and Technologies (SABITA), Regenerative and Restorative Medical Research Center (REMER), 34810, Istanbul, Turkey
| |
Collapse
|
173
|
D’Amore VM, Conflitti P, Marinelli L, Limongelli V. Minute-timescale free-energy calculations reveal a pseudo-active state in the adenosine A 2A receptor activation mechanism. Chem 2024; 10:3678-3698. [PMID: 40191447 PMCID: PMC11965979 DOI: 10.1016/j.chempr.2024.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 07/11/2024] [Accepted: 08/08/2024] [Indexed: 04/09/2025]
Abstract
G protein-coupled receptors (GPCRs) are membrane proteins targeted by over one-third of marketed drugs. Understanding their activation mechanism is essential for precise regulation of drug pharmacological response. In this work, we elucidate the conformational landscape of the adenosine A2A receptor (A2AR) activation mechanism in its basal apo form and under different ligand-bound conditions through minute-timescale free-energy calculations. We identified a pseudo-active state (pAs) of the A2AR apo form, stabilized by specific "microswitch" residues, including a salt bridge established between the conserved residues R5.66 and E6.30. The pAs enables A2AR to couple with Gs protein upon rearrangement of the intracellular end of transmembrane helix 6, providing unprecedented structural insights into receptor function and signaling dynamics. Our simulation protocol is versatile and can be adapted to study the activation of any GPCRs, potentially making it a valuable tool for drug design and "biased signaling" studies.
Collapse
Affiliation(s)
- Vincenzo Maria D’Amore
- Dipartimento di Farmacia, Università degli Studi di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Paolo Conflitti
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), via G. Buffi 13, CH-6900 Lugano, Switzerland
| | - Luciana Marinelli
- Dipartimento di Farmacia, Università degli Studi di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Vittorio Limongelli
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), via G. Buffi 13, CH-6900 Lugano, Switzerland
| |
Collapse
|
174
|
Kosenkov AM, Mal'tseva VN, Maiorov SA, Gaidin SG. The role of the endocannabinoid system in the pathogenesis and treatment of epilepsy. Rev Neurosci 2024:revneuro-2024-0114. [PMID: 39660979 DOI: 10.1515/revneuro-2024-0114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/15/2024] [Indexed: 12/12/2024]
Abstract
Epilepsy is a group of chronic neurological brain disorders characterized by recurrent spontaneous unprovoked seizures, which are accompanied by significant neurobiological, cognitive, and psychosocial impairments. With a global prevalence of approximately 0.5-1 % of the population, epilepsy remains a serious public health concern. Despite the development and widespread use of over 20 anticonvulsant drugs, around 30 % of patients continue to experience drug-resistant seizures, leading to a substantial reduction in quality of life and increased mortality risk. Given the limited efficacy of current treatments, exploring new therapeutic approaches is critically important. In recent years, Gi-protein-coupled receptors, particularly cannabinoid receptors CB1 and CB2, have garnered increasing attention as promising targets for the treatment seizures and prevention of epilepsy. Emerging evidence suggests a significant role of the cannabinoid system in modulating neuronal activity and protecting against hyperexcitability, underscoring the importance of further research in this area. This review provides up-to-date insights into the pathogenesis and treatment of epilepsy, with a special focus on the role of the cannabinoid system, highlighting the need for continued investigation to develop more effective therapeutic strategies.
Collapse
Affiliation(s)
- Artem M Kosenkov
- Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institute of Cell Biophysics of the Russian Academy of Sciences, 142290 Pushchino, Russian Federation
| | - Valentina N Mal'tseva
- Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institute of Cell Biophysics of the Russian Academy of Sciences, 142290 Pushchino, Russian Federation
| | - Sergei A Maiorov
- Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institute of Cell Biophysics of the Russian Academy of Sciences, 142290 Pushchino, Russian Federation
| | - Sergei G Gaidin
- Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Institute of Cell Biophysics of the Russian Academy of Sciences, 142290 Pushchino, Russian Federation
| |
Collapse
|
175
|
Dhamotharan K, Korn SM, Wacker A, Becker MA, Günther S, Schwalbe H, Schlundt A. A core network in the SARS-CoV-2 nucleocapsid NTD mediates structural integrity and selective RNA-binding. Nat Commun 2024; 15:10656. [PMID: 39653699 PMCID: PMC11628620 DOI: 10.1038/s41467-024-55024-0] [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: 06/17/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024] Open
Abstract
The SARS-CoV-2 nucleocapsid protein is indispensable for viral RNA genome processing. Although the N-terminal domain (NTD) is suggested to mediate specific RNA-interactions, high-resolution structures with viral RNA are still lacking. Available hybrid structures of the NTD with ssRNA and dsRNA provide valuable insights; however, the precise mechanism of complex formation remains elusive. Similarly, the molecular impact of nucleocapsid NTD mutations that have emerged since 2019 has not yet been fully explored. Using crystallography and solution NMR, we investigate how NTD mutations influence structural integrity and RNA-binding. We find that both features rely on a core network of residues conserved in Betacoronaviruses, crucial for protein stability and communication among flexible loop-regions that facilitate RNA-recognition. Our comprehensive structural analysis demonstrates that contacts within this network guide selective RNA-interactions. We propose that the core network renders the NTD evolutionarily robust in stability and plasticity for its versatile RNA processing roles.
Collapse
Affiliation(s)
- Karthikeyan Dhamotharan
- Institute for Molecular Biosciences, Goethe University, Frankfurt, Germany
- Center for Biomolecular Magnetic Resonance (BMRZ), Goethe University, Frankfurt, Germany
| | - Sophie M Korn
- Institute for Molecular Biosciences, Goethe University, Frankfurt, Germany.
- Center for Biomolecular Magnetic Resonance (BMRZ), Goethe University, Frankfurt, Germany.
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA.
| | - Anna Wacker
- Center for Biomolecular Magnetic Resonance (BMRZ), Goethe University, Frankfurt, Germany
- Institute for Organic Chemistry and Chemical Biology, Goethe University, Frankfurt, Germany
| | - Matthias A Becker
- Center for Biomolecular Magnetic Resonance (BMRZ), Goethe University, Frankfurt, Germany
- Institute for Organic Chemistry and Chemical Biology, Goethe University, Frankfurt, Germany
| | - Sebastian Günther
- Center for Free-Electron Laser Science CFEL, Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, Hamburg, Germany
| | - Harald Schwalbe
- Center for Biomolecular Magnetic Resonance (BMRZ), Goethe University, Frankfurt, Germany
- Institute for Organic Chemistry and Chemical Biology, Goethe University, Frankfurt, Germany
| | - Andreas Schlundt
- Institute for Molecular Biosciences, Goethe University, Frankfurt, Germany.
- Center for Biomolecular Magnetic Resonance (BMRZ), Goethe University, Frankfurt, Germany.
- Institute of Biochemistry, University of Greifswald, Greifswald, Germany.
| |
Collapse
|
176
|
Peshenko IV, Olshevskaya EV, Dizhoor AM. Calcium-sensor proteins but not bicarbonate ion activate retinal photoreceptor membrane guanylyl cyclase in photoreceptors. Front Mol Neurosci 2024; 17:1509366. [PMID: 39717564 PMCID: PMC11663931 DOI: 10.3389/fnmol.2024.1509366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/20/2024] [Indexed: 12/25/2024] Open
Abstract
Retinal membrane guanylyl cyclase (RetGC), regulated by guanylyl cyclase activating proteins (GCAPs) via negative calcium-feedback, is one of the most critically important enzymes in vertebrate rod and cone physiology, enabling their sensitivity to light. It was also reported that, similarly to olfactory receptor guanylyl cyclase, bicarbonate anion directly stimulates RetGC activity in photoreceptors as a novel phototransduction-linked regulating factor. We directly tested whether or not RetGC is a bicarbonate-activated enzyme using recombinant human RetGC expressed in HEK293 cells and the native RetGC in mouse retinas. Whereas RetGC in all cases was activated by GCAPs, we found no evidence indicating that bicarbonate can produce direct stimulating effect on RetGC catalytic activity, either basal or GCAP-activated, even at concentrations as high as 100 mM. Instead, near-physiological concentrations of bicarbonate only slightly reduced RetGC activity, whereas concentrations substantially exceeding physiological levels caused a more pronounced reduction of RetGC activity measured in mouse retinas. Our results argue that photoreceptor guanylyl cyclase is not a bicarbonate-stimulated enzyme and rule out the possibility that effects of bicarbonate on photoreceptor physiology are mediated by a direct stimulation of retinal guanylyl cyclase by HCO3 -.
Collapse
Affiliation(s)
- Igor V. Peshenko
- Pennsylvania College of Optometry, Salus at Drexel University, Elkins Park, PA, United States
| | - Elena V. Olshevskaya
- Pennsylvania College of Optometry, Salus at Drexel University, Elkins Park, PA, United States
| | - Alexander M. Dizhoor
- Pennsylvania College of Optometry, Salus at Drexel University, Elkins Park, PA, United States
- Department of Neurobiology and Anatomy, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
177
|
Murali S, Aradhyam GK. Divergent roles of DRY and NPxxY motifs in selective activation of downstream signalling by the apelin receptor. Biochem J 2024; 481:1707-1722. [PMID: 39513765 DOI: 10.1042/bcj20240320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/24/2024] [Accepted: 11/08/2024] [Indexed: 11/15/2024]
Abstract
G protein-coupled receptors (GPCRs) serve as critical communication hubs, translating a wide range of extracellular signals into intracellular responses that govern numerous physiological processes. In class-A GPCRs, conserved motifs mediate conformational changes of the active states of the receptor, and signal transduction is achieved by selectively binding to Gα proteins and/or adapter protein, arrestin. Apelin receptor (APJR) is a class-A GPCR that regulates a wide range of intracellular signalling cascades in response to apelin and elabela peptide ligands. Understanding how conserved motifs within APJR mediate activation and signal specificity remains unexplored. This study focuses on the functional roles of the DRY and NPxxY motifs within APJR by analyzing their impact on downstream signaling pathways across the receptor's conformational ensembles. Our findings provide compelling evidence that mutations within the conserved DRY and NPxxY motifs of APJR significantly alter its conformational preferences where modification of DRY motif leads to abrogation of G-protein coupling and mutation of NPxxY motif causing abolition of β-arrestin-2 recruitment. These observations shed light on the importance of these motifs in APJR activation and its potential for functional selectivity, highlighting the role of DRY/NPxxY as conformational switches of APJR signalling.
Collapse
Affiliation(s)
- Subhashree Murali
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biological Sciences, Indian Institute of Technology Madras, Chennai, India
| | - Gopala Krishna Aradhyam
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biological Sciences, Indian Institute of Technology Madras, Chennai, India
| |
Collapse
|
178
|
Shinozuka K, Jerotic K, Mediano P, Zhao AT, Preller KH, Carhart-Harris R, Kringelbach ML. Synergistic, multi-level understanding of psychedelics: three systematic reviews and meta-analyses of their pharmacology, neuroimaging and phenomenology. Transl Psychiatry 2024; 14:485. [PMID: 39632810 PMCID: PMC11618481 DOI: 10.1038/s41398-024-03187-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 11/13/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
Serotonergic psychedelics induce altered states of consciousness and have shown potential for treating a variety of neuropsychiatric disorders, including depression and addiction. Yet their modes of action are not fully understood. Here, we provide a novel, synergistic understanding of psychedelics arising from systematic reviews and meta-analyses of three hierarchical levels of analysis: (1) subjective experience (phenomenology), (2) neuroimaging and (3) molecular pharmacology. Phenomenologically, medium and high doses of LSD yield significantly higher ratings of visionary restructuralisation than psilocybin on the 5-dimensional Altered States of Consciousness Scale. Our neuroimaging results reveal that, in general, psychedelics significantly strengthen between-network functional connectivity (FC) while significantly diminishing within-network FC. Pharmacologically, LSD induces significantly more inositol phosphate formation at the 5-HT2A receptor than DMT and psilocin, yet there are no significant between-drug differences in the selectivity of psychedelics for the 5-HT2A, 5-HT2C, or D2 receptors, relative to the 5-HT1A receptor. Our meta-analyses link DMT, LSD, and psilocybin to specific neural fingerprints at each level of analysis. The results show a highly non-linear relationship between these fingerprints. Overall, our analysis highlighted the high heterogeneity and risk of bias in the literature. This suggests an urgent need for standardising experimental procedures and analysis techniques, as well as for more research on the emergence between different levels of psychedelic effects.
Collapse
Affiliation(s)
- Kenneth Shinozuka
- Centre for Eudaimonia and Human Flourishing, Linacre College, University of Oxford, Oxford, UK.
- Department of Psychiatry, University of Oxford, Oxford, UK.
- Oxford Mathematics of Consciousness and Applications Network (OMCAN), University of Oxford, Oxford, UK.
| | - Katarina Jerotic
- Centre for Eudaimonia and Human Flourishing, Linacre College, University of Oxford, Oxford, UK
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Pedro Mediano
- Centre for Eudaimonia and Human Flourishing, Linacre College, University of Oxford, Oxford, UK
- Department of Computing, Imperial College London, London, UK
| | - Alex T Zhao
- Department of Statistics and Data Science (Alumnus), The Wharton School, University of Pennsylvania, Philadelphia, PA, USA
| | - Katrin H Preller
- Departments of Psychiatry, Neuroscience, and Psychology, Yale University, New Haven, CT, USA
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Robin Carhart-Harris
- Centre for Psychedelic Research, Imperial College London, London, UK
- Department of Neurology, University of California, San Francisco, CA, USA
- Department of Neurology, Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Morten L Kringelbach
- Centre for Eudaimonia and Human Flourishing, Linacre College, University of Oxford, Oxford, UK
- Department of Psychiatry, University of Oxford, Oxford, UK
- Department of Clinical Medicine, Center for Music in the Brain, Aarhus University, Aarhus, Denmark
| |
Collapse
|
179
|
Dinsmore TC, Liu J, Miao J, Ünsal Ö, Sürmeli D, Beinborn M, Lin YS, Kumar K. Potent and Protease Resistant Azapeptide Agonists of the GLP-1 and GIP Receptors. Angew Chem Int Ed Engl 2024; 63:e202410237. [PMID: 39151024 DOI: 10.1002/anie.202410237] [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: 05/30/2024] [Revised: 07/17/2024] [Accepted: 08/13/2024] [Indexed: 08/18/2024]
Abstract
The gut-derived peptide hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) play important physiological roles including glucose homeostasis and appetite suppression. Stabilized agonists of the GLP-1 receptor (GLP-1R) and dual agonists of GLP-1R and GIP receptor (GIPR) for the management of type 2 diabetes and obesity have generated widespread enthusiasm and have become blockbuster drugs. These therapeutics are refractory to the action of dipeptidyl peptidase-4 (DPP4), that catalyzes rapid removal of the two N-terminal residues of the native peptides, in turn severely diminishing their activity profiles. Here we report that a single atom change from carbon to nitrogen in the backbone of the entire peptide makes them refractory to DPP4 action while still retaining full potency and efficacy at their respective receptors. This was accomplished by use of aza-amino acids, that are bioisosteric replacements for α-amino acids that perturb the structural backbone and local side chain conformations. Molecular dynamics simulations reveal that aza-amino acid can populate the same conformational space that GLP-1 adopts when bound to the GLP-1R. The insertion of an aza-amino acid at the second position from the N-terminus in semaglutide and in a dual agonist of GLP-1R and GIPR further demonstrates its capability as a viable alternative to current DPP4 resistance strategies while offering additional structural variation that may influence downstream signaling.
Collapse
Affiliation(s)
- Tristan C Dinsmore
- Department of Chemistry, Tufts University, 02155, Medford, Massachusetts, USA
| | - Jamie Liu
- Department of Chemistry, Tufts University, 02155, Medford, Massachusetts, USA
| | - Jiayuan Miao
- Department of Chemistry, Tufts University, 02155, Medford, Massachusetts, USA
| | - Özge Ünsal
- Department of Chemistry, Tufts University, 02155, Medford, Massachusetts, USA
| | - Damla Sürmeli
- Department of Chemistry, Tufts University, 02155, Medford, Massachusetts, USA
| | - Martin Beinborn
- Department of Chemistry, Tufts University, 02155, Medford, Massachusetts, USA
- Molecular Pharmacology Research Center, Tufts Medical Center, 02111, Boston, Massachusetts, USA
| | - Yu-Shan Lin
- Department of Chemistry, Tufts University, 02155, Medford, Massachusetts, USA
| | - Krishna Kumar
- Department of Chemistry, Tufts University, 02155, Medford, Massachusetts, USA
- Department of Biomedical Engineering, Tufts University, 02155, Medford, Massachusetts, USA
- Cancer Center, Tufts Medical Center, 02111, Boston, Massachusetts, USA
| |
Collapse
|
180
|
Raza SHA, Zhong R, Yu X, Zhao G, Wei X, Lei H. Advances of Predicting Allosteric Mechanisms Through Protein Contact in New Technologies and Their Application. Mol Biotechnol 2024; 66:3385-3397. [PMID: 37957479 DOI: 10.1007/s12033-023-00951-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/12/2023] [Indexed: 11/15/2023]
Abstract
Allostery is an intriguing phenomenon wherein the binding activity of a biological macromolecule is modulated via non-canonical binding site, resulting in synchronized functional changes. The mechanics underlying allostery are relatively complex and this review is focused on common methodologies used to study allostery, such as X-ray crystallography, NMR spectroscopy, and HDXMS. Different methodological approaches are used to generate data in different scenarios. For example, X-ray crystallography provides high-resolution structural information, NMR spectroscopy offers dynamic insights into allosteric interactions in solution, and HDXMS provides information on protein dynamics. The residue transition state (RTS) approach has emerged as a critical tool in understanding the energetics and conformational changes associated with allosteric regulation. Allostery has significant implications in drug discovery, gene transcription, disease diagnosis, and enzyme catalysis. Enzymes' catalytic activity can be modulated by allosteric regulation, offering opportunities to develop novel therapeutic alternatives. Understanding allosteric mechanisms associated with infectious organisms like SARS-CoV and bacterial pathogens can aid in the development of new antiviral drugs and antibiotics. Allosteric mechanisms are crucial in the regulation of a variety of signal transduction and cell metabolism pathways, which in turn govern various cellular processes. Despite progress, challenges remain in identifying allosteric sites and characterizing their contribution to a variety of biological processes. Increased understanding of these mechanisms can help develop allosteric systems specifically designed to modulate key biological mechanisms, providing novel opportunities for the development of targeted therapeutics. Therefore, the current review aims to summarize common methodologies that are used to further our understanding of allosteric mechanisms. In conclusion, this review provides insights into the methodologies used for the study of allostery, its applications in in silico modeling, the mechanisms underlying antibody allostery, and the ongoing challenges and prospects in advancing our comprehension of this intriguing phenomenon.
Collapse
Affiliation(s)
- Sayed Haidar Abbas Raza
- Guangdong Provincial Key Laboratory of Food Quality and Safety/Nation-Local Joint Engineering Research Center for Machining and Safety of Livestock and Poultry Products, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan, 512005, China
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Ruimin Zhong
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan, 512005, China
| | - Xiaoting Yu
- Guangdong Provincial Key Laboratory of Food Quality and Safety/Nation-Local Joint Engineering Research Center for Machining and Safety of Livestock and Poultry Products, South China Agricultural University, Guangzhou, 510642, China
| | - Gang Zhao
- Guangdong Provincial Key Laboratory of Food Quality and Safety/Nation-Local Joint Engineering Research Center for Machining and Safety of Livestock and Poultry Products, South China Agricultural University, Guangzhou, 510642, China
| | - Xiaoqun Wei
- Guangdong Provincial Key Laboratory of Food Quality and Safety/Nation-Local Joint Engineering Research Center for Machining and Safety of Livestock and Poultry Products, South China Agricultural University, Guangzhou, 510642, China
| | - Hongtao Lei
- Guangdong Provincial Key Laboratory of Food Quality and Safety/Nation-Local Joint Engineering Research Center for Machining and Safety of Livestock and Poultry Products, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, 510642, China.
- Licheng Detection and Certification Group Co., Ltd., Zhongshan, 528403, Guangdong, China.
| |
Collapse
|
181
|
Liu X, Cherepanov S, Abouzari M, Zuko A, Yang S, Sayadi J, Jia X, Terao C, Sasaki T, Yokoyama S. R150S mutation in the human oxytocin receptor: Gain-of-function effects and implication in autism spectrum disorder. Peptides 2024; 182:171301. [PMID: 39395443 DOI: 10.1016/j.peptides.2024.171301] [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: 02/29/2024] [Revised: 09/18/2024] [Accepted: 10/05/2024] [Indexed: 10/14/2024]
Abstract
This study investigates the rs547238576 (R150S) missense variant in the oxytocin receptor (OXTR) gene, previously observed through screening of rare variants in Japanese individuals with autism spectrum disorders (ASD). Contrary to the anticipated loss-of-function, R150S exhibits gain-of-function effects, enhancing oxytocin (OXT) sensitivity, ligand-binding affinity, and OXT-induced Ca2+ mobilization in vitro. This suggests R150S may alter OXT signaling, potentially contributing to the excitatory/inhibitory imbalance seen in ASD and other psychiatric disorders. Our findings underscore the significance of genetic variations in OXTR on functional activity and highlight the necessity for population-specific genetic study and in vitro analysis to elucidate genetic susceptibilities to neuropsychiatric conditions.
Collapse
Affiliation(s)
- Xiaoxi Liu
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Stanislav Cherepanov
- Research Center for Child Mental Development, Kanazawa University, Kanazawa, Kanazawa, Ishikawa, Japan; Division of Socio-Cognitive-Neuroscience, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Kanazawa, Japan; Institute for Functional Genomics, French National Centre for Scientific Research, Montpellier, Occitanie, France
| | - Mehdi Abouzari
- Department of Otolaryngology-Head and Neck Surgery, University of California, Irvine, CA 92697, USA
| | - Amila Zuko
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, the Netherlands
| | - Shu Yang
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, USA
| | - Jamasb Sayadi
- Stanford University School of Medicine, Palo Alto, CA, USA
| | - Xiaoyuan Jia
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan; The Department of Applied Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Tsukasa Sasaki
- Department of Physical and Health Education, Graduate School of Education, The University of Tokyo, Tokyo, Japan
| | - Shigeru Yokoyama
- Research Center for Child Mental Development, Kanazawa University, Kanazawa, Kanazawa, Ishikawa, Japan; Division of Socio-Cognitive-Neuroscience, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Kanazawa, Japan.
| |
Collapse
|
182
|
Yang Z, Li H, Wu HY, Zhou Y, Du JX, Hu ZX. Omega-3 polyunsaturated fatty acids alleviate hyperuricemic nephropathy by inhibiting renal pyroptosis through GPR120. Biochem Pharmacol 2024; 230:116575. [PMID: 39396646 DOI: 10.1016/j.bcp.2024.116575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Hyperuricemic nephropathy (HN) is characterized by increased serum uric acid levels that incite renal inflammation. While omega-3 polyunsaturated fatty acids (PUFAs) are known for their anti-inflammatory properties, their impact on HN remains unclear. This study explored the effects of omega-3 PUFAs, specifically docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), on HN. Using a mouse model induced by adenine and potassium oxonate, we treated HN mice with DHA, EPA, or both for four weeks. The results showed that omega-3 PUFAs significantly reduced serum uric acid levels and improved kidney function, with DHA, EPA, and their combination showing similar efficacy. Transcriptome sequencing and further analysis revealed that these fatty acids alleviate renal pyroptosis by reducing key markers such as NOD-like receptor pyrin containing 3 (NLRP3), cleaved gasdermin-D, caspase-1, and interleukin-1β. To further investigate the underlying mechanism, we focused on G-protein coupled receptor 120 (GPR120), a receptor activated by DHA. The use of a GPR120 antagonist (AH7614) partially blocked DHA's effects, while the agonist (TUG891) mimicked its anti-pyroptotic actions. Co-immunoprecipitation assays showed that DHA activates GPR120, leading to its internalization and interaction with β-arrestin2, ultimately inhibiting NLRP3 inflammasome formation and reducing inflammation. Overall, omega-3 PUFAs, particularly through GPR120 activation, appear to protect against renal inflammation in HN by modulating the NLRP3/caspase-1/GSDMD pathway.
Collapse
Affiliation(s)
- Zhi Yang
- Department of Nephrology, West China Hospital, Sichuan University, Guoxue Alley 37#, Chengdu 610041, Sichuan, PR China
| | - Hao Li
- Department of Nephrology, West China Hospital, Sichuan University, Guoxue Alley 37#, Chengdu 610041, Sichuan, PR China
| | - Hong-Yan Wu
- Department of Nephrology, West China Hospital, Sichuan University, Guoxue Alley 37#, Chengdu 610041, Sichuan, PR China
| | - Yi Zhou
- Department of Nephrology, West China Hospital, Sichuan University, Guoxue Alley 37#, Chengdu 610041, Sichuan, PR China
| | - Jing-Xue Du
- Department of Nephrology, West China Hospital, Sichuan University, Guoxue Alley 37#, Chengdu 610041, Sichuan, PR China
| | - Zhang-Xue Hu
- Department of Nephrology, West China Hospital, Sichuan University, Guoxue Alley 37#, Chengdu 610041, Sichuan, PR China.
| |
Collapse
|
183
|
Felline A, Bellucci L, Vezzi V, Ambrosio C, Cotecchia S, Fanelli F. Structural plasticity of arrestin-G protein coupled receptor complexes as a molecular determinant of signaling. Int J Biol Macromol 2024; 283:137217. [PMID: 39515728 DOI: 10.1016/j.ijbiomac.2024.137217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/27/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
G protein coupled receptors (GPCRs) are critically regulated by arrestins. In this study, high-resolution data was combined with molecular dynamics simulations to infer the determinants of β-arrestin 1 (βarr1)-GPCR coupling, using the V2 vasopressin receptor (V2R) as a model system. The study highlighted the extremely high plasticity of βarr1-GPCR complexes, dependent on receptor type, state, and membrane environment. The multiple functions of receptor-bound βarr1 are likely determined by the interplay of intrinsic flexibility and collective motions both as a bi-domain protein and as a whole. The two major collective motions of the whole βarr1, consisting in rotation parallel to the membrane plane and inclination with respect to the receptor main axis, are distinctly linked to the two intermolecular interfaces involved in tail and core interactions. The intermolecular dynamic coupling between βarr1 and V2R depends on the allosteric effect of the agonist arginine-vasopressin (AVP). In the absence of AVP the dynamic coupling concerns only tail interactions, while in the presence of AVP it involves both tail and core interactions. This suggests that constitutive and agonist-induced arrestin-receptor dynamic coupling is linked to distinct arrestin functions.
Collapse
Affiliation(s)
- Angelo Felline
- Dipartimento di Scienze della Vita, Università di Modena e Reggio Emilia, via Campi 103, 41125 Modena, Italy
| | - Luca Bellucci
- NEST, Istituto Nanoscienze-CNR, Piazza San Silvestro 12, 56127 Pisa, Italy
| | - Vanessa Vezzi
- Istituto Superiore di Sanità, V.le Regina Elena, 299 00161 Roma, Italy
| | - Caterina Ambrosio
- Istituto Superiore di Sanità, V.le Regina Elena, 299 00161 Roma, Italy
| | - Susanna Cotecchia
- Dipartimento di Bioscienze, Biotecnologie e Ambiente, Università di Bari, via Orabona 4, 70125 Bari, Italy
| | - Francesca Fanelli
- Dipartimento di Scienze della Vita, Università di Modena e Reggio Emilia, via Campi 103, 41125 Modena, Italy.
| |
Collapse
|
184
|
Ahmed MR, Inayathullah M, Morton M, Pothineni VR, Kim K, Ahmed MS, Babar MM, Rajadas J. Intranasal delivery of liposome encapsulated flavonoids ameliorates l-DOPA induced dyskinesia in hemiparkinsonian mice. Biomaterials 2024; 311:122680. [PMID: 38959534 DOI: 10.1016/j.biomaterials.2024.122680] [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: 01/02/2024] [Revised: 04/25/2024] [Accepted: 06/23/2024] [Indexed: 07/05/2024]
Abstract
In the present study, we explored the development of a novel noninvasive liposomal drug delivery material for use in intranasal drug delivery applications in human diseases. We used drug entrapment into liposomal nanoparticle assembly to efficiently deliver the drugs to the nasal mucosa to be delivered to the brain. The naturally occurring flavonoid 7,8-dihydroxyflavone (7,8-DHF) has previously been shown to have beneficial effects in ameliorating Parkinson's disease (PD). We used both naturally occurring 7,8-DHF and the chemically modified form of DHF, the DHF-ME, to be used as a drug candidate for the treatment of PD and l-DOPA induced dyskinesia (LID), which is the debilitating side effect of l-DOPA therapy in PD. The ligand-protein interaction behavior for 7,8-DHF and 6,7-DHF-ME was found to be more effective with molecular docking and molecular stimulation studies of flavonoid compounds with TrkB receptor. Our study showed that 7,8-DHF delivered via intranasal route using a liposomal formulation ameliorated LID in hemiparkinsonian mice model when these mice were chronically administered with l-DOPA, which is the only current medication for relieving the clinical symptoms of PD. The present study also demonstrated that apart from reducing the LID, 7,8-DHF delivery directly to the brain via the intranasal route also corrected some long-term signaling adaptations involving ΔFosB and α Synuclein in the brain of dopamine (DA) depleted animals.
Collapse
Affiliation(s)
- Mohamed Rafiuddin Ahmed
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA
| | - Mohammed Inayathullah
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA
| | - Mithya Morton
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA; Children's Hospital of Orange County - UC Irvine School of Medicine, Department of Pediatrics, 505 S. Main St., Suite #525, Orange, CA, 92868, USA
| | - Venkata Raveendra Pothineni
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA
| | - Kwangmin Kim
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA; Department of Physiology, Gachon University College of Medicine, Incheon, 21999, South Korea
| | - Mohamed Sohail Ahmed
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, MCN 1161 21st Ave S. Nashville TN 37232, USA
| | - Mustafeez Mujtaba Babar
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA
| | - Jayakumar Rajadas
- Advanced Drug Delivery and Regenerative Biomaterials Laboratory of Cardiovascular Institute/ Pulmonary and Critical Care, Department of Medicine, Stanford University, 1050 Arastradero Road, Palo Alto, CA, 94304, USA.
| |
Collapse
|
185
|
Barreto CA, Moreira IS. An atomic look at the interface of GHSR and its partners. Comput Struct Biotechnol J 2024; 23:4242-4251. [PMID: 39660221 PMCID: PMC11629268 DOI: 10.1016/j.csbj.2024.11.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/12/2024] Open
Abstract
G protein-coupled receptors (GPCRs) regulate cellular activity by transducing external signals and selectively coupling them to intracellular partners. Ghrelin receptor (GHSR) has garnered significant interest over the past decade owing to its diverse functional roles. In this study, we simulated five distinct GHSR-partner complexes, including Gq, Gi, and arrestin in two conformational states, to investigate the structural determinants of partner coupling. Interface and contact analyses revealed conserved interaction sites and novel interactions that were specific to each partner family. Molecular dynamics simulations provided insights into GHSR conformational dynamics, highlighting notable differences in key structural regions across complexes, such as the TM5 bulge. Our findings underscore the structural diversity of GHSR coupling mechanisms and contribute to a deeper understanding of their functional versatility.
Collapse
Affiliation(s)
- Carlos A.V. Barreto
- PhD Programme in Experimental Biology and Biomedicine, Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Casa Costa Alemão, Coimbra 3030–789 , Portugal
- CNC - Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004–504, Portugal
| | - Irina S. Moreira
- CNC - Center for Neuroscience and Cell Biology, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004–504, Portugal
- Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, Coimbra 3000–456, Portugal
| |
Collapse
|
186
|
Qi F, Chen X, Wang J, Niu X, Li S, Huang S, Ran X. Genome-wide characterization of structure variations in the Xiang pig for genetic resistance to African swine fever. Virulence 2024; 15:2382762. [PMID: 39092797 PMCID: PMC11299630 DOI: 10.1080/21505594.2024.2382762] [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: 10/09/2023] [Revised: 05/07/2024] [Accepted: 07/12/2024] [Indexed: 08/04/2024] Open
Abstract
African swine fever (ASF) is a rapidly fatal viral haemorrhagic fever in Chinese domestic pigs. Although very high mortality is observed in pig farms after an ASF outbreak, clinically healthy and antibody-positive pigs are found in those farms, and viral detection is rare from these pigs. The ability of pigs to resist ASF viral infection may be modulated by host genetic variations. However, the genetic basis of the resistance of domestic pigs against ASF remains unclear. We generated a comprehensive set of structural variations (SVs) in a Chinese indigenous Xiang pig with ASF-resistant (Xiang-R) and ASF-susceptible (Xiang-S) phenotypes using whole-genome resequencing method. A total of 53,589 nonredundant SVs were identified, with an average of 25,656 SVs per individual in the Xiang pig genome, including insertion, deletion, inversion and duplication variations. The Xiang-R group harboured more SVs than the Xiang-S group. The F-statistics (FST) was carried out to reveal genetic differences between two populations using the resequencing data at each SV locus. We identified 2,414 population-stratified SVs and annotated 1,152 Ensembl genes (including 986 protein-coding genes), in which 1,326 SVs might disturb the structure and expression of the Ensembl genes. Those protein-coding genes were mainly enriched in the Wnt, Hippo, and calcium signalling pathways. Other important pathways associated with the ASF viral infection were also identified, such as the endocytosis, apoptosis, focal adhesion, Fc gamma R-mediated phagocytosis, junction, NOD-like receptor, PI3K-Akt, and c-type lectin receptor signalling pathways. Finally, we identified 135 candidate adaptive genes overlapping 166 SVs that were involved in the virus entry and virus-host cell interactions. The fact that some of population-stratified SVs regions detected as selective sweep signals gave another support for the genetic variations affecting pig resistance against ASF. The research indicates that SVs play an important role in the evolutionary processes of Xiang pig adaptation to ASF infection.
Collapse
Affiliation(s)
- Fenfang Qi
- Institute of Agro-Bioengineering, Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences, College of Animal Science, Guizhou University, Guiyang, Guizhou Province, China
| | - Xia Chen
- Institute of Agro-Bioengineering, Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences, College of Animal Science, Guizhou University, Guiyang, Guizhou Province, China
| | - Jiafu Wang
- Institute of Agro-Bioengineering, Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences, College of Animal Science, Guizhou University, Guiyang, Guizhou Province, China
| | - Xi Niu
- Institute of Agro-Bioengineering, Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences, College of Animal Science, Guizhou University, Guiyang, Guizhou Province, China
| | - Sheng Li
- Institute of Agro-Bioengineering, Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences, College of Animal Science, Guizhou University, Guiyang, Guizhou Province, China
| | - Shihui Huang
- Institute of Agro-Bioengineering, Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences, College of Animal Science, Guizhou University, Guiyang, Guizhou Province, China
| | - Xueqin Ran
- Institute of Agro-Bioengineering, Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), College of Life Sciences, College of Animal Science, Guizhou University, Guiyang, Guizhou Province, China
| |
Collapse
|
187
|
Li F, Liu R, Li W, Xie M, Qin S. Synchrotron Radiation: A Key Tool for Drug Discovery. Bioorg Med Chem Lett 2024; 114:129990. [PMID: 39406298 DOI: 10.1016/j.bmcl.2024.129990] [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: 07/18/2024] [Revised: 09/22/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024]
Abstract
Synchrotron radiation is extensively utilized in the domains of materials science, physical chemistry, and life science, resulting from its high intensity, exceptional monochromaticity, superior collimation, and broad wave spectrum. This top-notch light source has also made significant contributions to the progress of biomedicine. The advancement of synchrotron radiation-based X-ray and protein crystallography technologies has created new prospects for drug discovery. These innovative techniques have opened up exciting avenues in the field. The investigation of protein crystal structures and the elucidation of the spatial configuration of biological macromolecules have revealed intricate details regarding the modes of protein binding. Furthermore, the screening of crystal polymorphs and ligands has laid the groundwork for rational drug modification and the improvement of drug physicochemical properties. As science and technology continue to advance, the techniques for analyzing structures using synchrotron radiation sources and the design of corresponding crystallographic beamline stations are undergoing continuous enhancement. These cutting-edge tools and facilities are expected to expedite the drug development process and rectify the current situation of a lack of targeted drugs.
Collapse
Affiliation(s)
- Fengcheng Li
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao 266112, China; Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
| | - Runze Liu
- Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
| | - Wenjun Li
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao 266112, China; Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
| | - Mingyuan Xie
- Institute of Advanced Science Facilities, Shenzhen, Guangdong 518107, China.
| | - Song Qin
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao 266112, China; Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China.
| |
Collapse
|
188
|
Zhang C, Nie Y, Xu B, Mu C, Tian GG, Li X, Cheng W, Zhang A, Li D, Wu J. Luteinizing Hormone Receptor Mutation (LHR N316S) Causes Abnormal Follicular Development Revealed by Follicle Single-Cell Analysis and CRISPR/Cas9. Interdiscip Sci 2024; 16:976-989. [PMID: 39150470 PMCID: PMC11512921 DOI: 10.1007/s12539-024-00646-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/17/2024]
Abstract
Abnormal interaction between granulosa cells and oocytes causes disordered development of ovarian follicles. However, the interactions between oocytes and cumulus granulosa cells (CGs), oocytes and mural granulosa cells (MGs), and CGs and MGs remain to be fully explored. Using single-cell RNA-sequencing (scRNA-seq), we determined the transcriptional profiles of oocytes, CGs and MGs in antral follicles. Analysis of scRNA-seq data revealed that CGs may regulate follicular development through the BMP15-KITL-KIT-PI3K-ARF6 pathway with elevated expression of luteinizing hormone receptor (LHR). Because internalization of the LHR is regulated by Arf6, we constructed LHRN316S mice by CRISPR/Cas9 to further explore mechanisms of follicular development and novel treatment strategies for female infertility. Ovaries of LHRN316S mice exhibited reduced numbers of corpora lutea and ovulation. The LHRN316S mice had a reduced rate of oocyte maturation in vitro and decreased serum progesterone levels. Mating LHRN316S female mice with ICR wild type male mice revealed that the infertility rate of LHRN316S mice was 21.4% (3/14). Litter sizes from LHRN316S mice were smaller than those from control wild type female mice. The oocytes from LHRN316S mice had an increased rate of maturation in vitro after progesterone administration in vitro. Furthermore, progesterone treated LHRN316S mice produced offspring numbers per litter equivalent to WT mice. These findings provide key insights into cellular interactions in ovarian follicles and provide important clues for infertility treatment.
Collapse
Affiliation(s)
- Chen Zhang
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
- Department of Hematology, Tangdu Hospital, Xi'an, 710032, China
| | - Yongqiang Nie
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Bufang Xu
- Department of Obstetrics and Gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chunlan Mu
- School of Basic Medical Sciences, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China
| | - Geng G Tian
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoyong Li
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Weiwei Cheng
- International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Aijun Zhang
- Department of Obstetrics and Gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Dali Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Ji Wu
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China.
- School of Basic Medical Sciences, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
189
|
Barnes CL, Salom D, Namitz KEW, Smith WC, Knutson BA, Cosgrove MS, Kiser PD, Calvert PD. Mechanisms of amphibian arrestin 1 self-association and dynamic distribution in retinal photoreceptors. J Biol Chem 2024; 300:107966. [PMID: 39510183 PMCID: PMC11652889 DOI: 10.1016/j.jbc.2024.107966] [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: 07/08/2024] [Revised: 10/15/2024] [Accepted: 10/26/2024] [Indexed: 11/15/2024] Open
Abstract
Visual arrestin 1 (Arr1) is an essential protein for termination of the light response in photoreceptors. While mammalian Arr1s form dimers and tetramers at physiological concentrations in vitro, oligomerization in other vertebrates has not been studied. Here we examine self-association of Arr1 from two amphibian species, Xenopus laevis (xArr1) and Ambystoma tigrinum (salArr1). Sedimentation velocity analytical ultracentrifugation showed that xArr1 and salArr1 oligomerization is limited to dimers. The KD for dimer formation was 53 μM for xArr1 and 44 μM for salArr1, similar to the 69 μM KD for bovine Arr1 (bArr1) dimers. Mutations of orthologous amino acids important for mammalian Arr1 oligomerization had no impact on xArr1 dimerization. Crystallography showed that the fold of xArr1 closely resembles that of bArr1 and crystal structures in different space groups revealed two potential xArr1 dimer forms: a symmetric dimer with a C-domain interface (CC dimer), resembling the bArr1 solution dimer, and an asymmetric dimer with an N-domain/C-domain interface. Mutagenesis of residues predicted to interact in either of these two dimer forms yielded modest reduction in dimer affinity, suggesting that the dimer interfaces compete or are not unique. Indeed, small-angle X-ray scattering and protein painting data were consistent with a symmetric anti-parallel solution dimer (AP dimer) distinct from the assemblies observed by crystallography. Finally, a computational model evaluating xArr1 binding to compartment-specific partners and partitioning based on heterogeneity of available cytoplasmic spaces shows that Arr1 distribution in dark-adapted photoreceptors is largely explained by the excluded volume effect together with tuning by oligomerization.
Collapse
Affiliation(s)
- Cassandra L Barnes
- Center for Vision Research and the Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
| | - David Salom
- Department of Ophthalmology, Gavin Herbert Eye Institute - Center for Translational Vision Research, University of California, Irvine, California, USA
| | - Kevin E W Namitz
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - W Clay Smith
- Department of Ophthalmology, University of Florida, Gainesville, Florida, USA
| | - Bruce A Knutson
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Michael S Cosgrove
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Philip D Kiser
- Department of Ophthalmology, Gavin Herbert Eye Institute - Center for Translational Vision Research, University of California, Irvine, California, USA; Department of Physiology & Biophysics, University of California, Irvine, California, USA; Research Service, VA Long Beach Medical Center, Long Beach, California, USA.
| | - Peter D Calvert
- Center for Vision Research and the Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, New York, USA.
| |
Collapse
|
190
|
Yang H, Yang J, Zheng X, Chen T, Zhang R, Chen R, Cao T, Zeng F, Liu Q. The Hippo Pathway in Breast Cancer: The Extracellular Matrix and Hypoxia. Int J Mol Sci 2024; 25:12868. [PMID: 39684583 DOI: 10.3390/ijms252312868] [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: 11/07/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
As one of the most prevalent malignant neoplasms among women globally, the optimization of therapeutic strategies for breast cancer has perpetually been a research hotspot. The tumor microenvironment (TME) is of paramount importance in the progression of breast cancer, among which the extracellular matrix (ECM) and hypoxia are two crucial factors. The alterations of these two factors are predominantly regulated by the Hippo signaling pathway, which promotes tumor invasiveness, metastasis, therapeutic resistance, and susceptibility. Hence, this review focuses on the Hippo pathway in breast cancer, specifically, how the ECM and hypoxia impact the biological traits and therapeutic responses of breast cancer. Moreover, the role of miRNAs in modulating ECM constituents was investigated, and hsa-miR-33b-3p was identified as a potential therapeutic target for breast cancer. The review provides theoretical foundations and potential therapeutic direction for clinical treatment strategies in breast cancer, with the aspiration of attaining more precise and effective treatment alternatives in the future.
Collapse
Affiliation(s)
- Hanyu Yang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Jiaxin Yang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Xiang Zheng
- School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Tianshun Chen
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Ranqi Zhang
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Rui Chen
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Tingting Cao
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
| | - Qiuyu Liu
- School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou 646000, China
- Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| |
Collapse
|
191
|
Batista Brochado AC, de Moraes JA, Rodrigues de Oliveira B, De Souza Lima VH, Mariano ED, Karande S, Romasco T, Leite PEC, Mourão CF, Gomes Alves G. Metabolic and Regulatory Pathways Involved in the Anticancer Activity of Perillyl Alcohol: A Scoping Review of In Vitro Studies. Cancers (Basel) 2024; 16:4003. [PMID: 39682189 PMCID: PMC11640718 DOI: 10.3390/cancers16234003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/25/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Perillyl alcohol (POH), a plant-derived compound, has demonstrated anti-tumor activity across various human cancers. Understanding the regulatory pathways through which POH exerts its effects is crucial for identifying new therapeutic opportunities and exploring potential drug repositioning strategies. Therefore, this scoping review aims to provide a comprehensive overview of the metabolic and regulatory pathways involved in the anticancer effects of POH, based on in vitro evidence. METHODS Following the PRISMA-ScR 2018 guidelines, a systematic search was conducted in the PUBMED, Web of Science, and Scopus databases. RESULTS A total of 39 studies were included, revealing that POH exerts its biological effects by modulating several pathways, including the regulation of cyclins, CDKs, and p21, thereby affecting cell cycle progression. It inhibits growth and promotes cell death by attenuating AKT phosphorylation, reducing PARP-1 activity, increasing caspase activity and the FAS receptor and its ligand FASL. Additionally, POH reduces ERK phosphorylation, inhibits RAS protein isoprenylation, and decreases Na/K-ATPase activity. CONCLUSIONS In conclusion, this review delineates the key regulatory pathways responsible for mediating the biological effects of POH in cancer.
Collapse
Affiliation(s)
- Ana Carolina Batista Brochado
- Post-Graduation Program in Science & Biotechnology, Institute of Biology, Fluminense Federal University, Niteroi 24220-900, Brazil
| | - Júlia Alves de Moraes
- Clinical Research Unit, Antonio Pedro University Hospital, Fluminense Federal University, Niteroi 24020-140, Brazil
| | - Bruna Rodrigues de Oliveira
- Clinical Research Unit, Antonio Pedro University Hospital, Fluminense Federal University, Niteroi 24020-140, Brazil
| | - Victor Hugo De Souza Lima
- Post-Graduation Program in Science & Biotechnology, Institute of Biology, Fluminense Federal University, Niteroi 24220-900, Brazil
| | | | - Sachin Karande
- Dental Research Division, Department of Periodontology and Oral Implantology, Fluminense Federal University, Niteroi 21941-617, Brazil
| | - Tea Romasco
- Division of Dental Research Administration, Tufts University School of Dental Medicine, Boston, MA 02111, USA
- Department of Medical, Oral and Biotechnological Sciences, Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Paulo Emilio Correa Leite
- Post-Graduation Program in Science & Biotechnology, Institute of Biology, Fluminense Federal University, Niteroi 24220-900, Brazil
| | - Carlos Fernando Mourão
- Department of Basic and Clinical Translational Sciences, Tufts University School of Dental Medicine, Boston, MA 02111, USA
| | - Gutemberg Gomes Alves
- Post-Graduation Program in Science & Biotechnology, Institute of Biology, Fluminense Federal University, Niteroi 24220-900, Brazil
- Clinical Research Unit, Antonio Pedro University Hospital, Fluminense Federal University, Niteroi 24020-140, Brazil
| |
Collapse
|
192
|
De Faveri C, Mattheisen JM, Sakmar TP, Coin I. Noncanonical Amino Acid Tools and Their Application to Membrane Protein Studies. Chem Rev 2024; 124:12498-12550. [PMID: 39509680 PMCID: PMC11613316 DOI: 10.1021/acs.chemrev.4c00181] [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: 03/01/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 11/15/2024]
Abstract
Methods rooted in chemical biology have contributed significantly to studies of integral membrane proteins. One recent key approach has been the application of genetic code expansion (GCE), which enables the site-specific incorporation of noncanonical amino acids (ncAAs) with defined chemical properties into proteins. Efficient GCE is challenging, especially for membrane proteins, which have specialized biogenesis and cell trafficking machinery and tend to be expressed at low levels in cell membranes. Many eukaryotic membrane proteins cannot be expressed functionally in E. coli and are most effectively studied in mammalian cell culture systems. Recent advances have facilitated broader applications of GCE for studies of membrane proteins. First, AARS/tRNA pairs have been engineered to function efficiently in mammalian cells. Second, bioorthogonal chemical reactions, including cell-friendly copper-free "click" chemistry, have enabled linkage of small-molecule probes such as fluorophores to membrane proteins in live cells. Finally, in concert with advances in GCE methodology, the variety of available ncAAs has increased dramatically, thus enabling the investigation of protein structure and dynamics by multidisciplinary biochemical and biophysical approaches. These developments are reviewed in the historical framework of the development of GCE technology with a focus on applications to studies of membrane proteins.
Collapse
Affiliation(s)
- Chiara De Faveri
- Faculty
of Life Science, Institute of Biochemistry, Leipzig University, Leipzig 04103, Germany
| | - Jordan M. Mattheisen
- Laboratory
of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York 10065, United States
- Tri-Institutional
PhD Program in Chemical Biology, New York, New York 10065, United States
| | - Thomas P. Sakmar
- Laboratory
of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York 10065, United States
| | - Irene Coin
- Faculty
of Life Science, Institute of Biochemistry, Leipzig University, Leipzig 04103, Germany
| |
Collapse
|
193
|
Dahlgren C, Forsman H, Sundqvist M, Björkman L, Mårtensson J. Signaling by neutrophil G protein-coupled receptors that regulate the release of superoxide anions. J Leukoc Biol 2024; 116:1334-1351. [PMID: 39056275 DOI: 10.1093/jleuko/qiae165] [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: 12/09/2023] [Revised: 06/18/2024] [Accepted: 07/24/2024] [Indexed: 07/28/2024] Open
Abstract
In human peripheral blood, the neutrophil granulocytes (neutrophils) are the most abundant white blood cells. These professional phagocytes are rapidly recruited from the bloodstream to inflamed tissues by chemotactic factors that signal danger. Neutrophils, which express many receptors that are members of the large family of G protein-coupled receptors (GPCRs), are critical for the elimination of pathogens and inflammatory insults, as well as for the resolution of inflammation leading to tissue repair. Danger signaling molecular patterns such as the N-formylated peptides that are formed during bacterial and mitochondrial protein synthesis and recognized by formyl peptide receptors (FPRs) and free fatty acids recognized by free fatty acid receptors (FFARs) regulate neutrophil functions. Short peptides and short-chain fatty acids activate FPR1 and FFA2R, respectively, while longer peptides and fatty acids activate FPR2 and GPR84, respectively. The activation profiles of these receptors include the release of reactive oxygen species (ROS) generated by the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. Activation of the oxidase and the production of ROS are processes that are regulated by proinflammatory mediators, including tumor necrosis factor α and granulocyte/macrophage colony-stimulating factor. The receptors have signaling and functional similarities, although there are also important differences, not only between the two closely related neutrophil FPRs, but also between the FPRs and the FFARs. In neutrophils, these receptors never walk alone, and additional mechanistic insights into the regulation of the GPCRs and the novel regulatory mechanisms underlying the activation of NADPH oxidase advance our understanding of the role of receptor transactivation in the regulation of inflammatory reactions.
Collapse
Affiliation(s)
- Claes Dahlgren
- Department of Rheumatology and Inflammation Research, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gulhedsgatan 10, Göteborg S-41346, Sweden
| | - Huamei Forsman
- Department of Rheumatology and Inflammation Research, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gulhedsgatan 10, Göteborg S-41346, Sweden
| | - Martina Sundqvist
- Department of Rheumatology and Inflammation Research, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gulhedsgatan 10, Göteborg S-41346, Sweden
| | - Lena Björkman
- Department of Rheumatology and Inflammation Research, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gulhedsgatan 10, Göteborg S-41346, Sweden
| | - Jonas Mårtensson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gulhedsgatan 10, Göteborg S-41346, Sweden
| |
Collapse
|
194
|
Upadhyay S, Kumar S, Singh VK, Tiwari R, Kumar A, Sundar S, Kumar R. Chemokines Signature and T Cell Dynamics in Leishmaniasis: Molecular insight and therapeutic application. Expert Rev Mol Med 2024; 27:1-55. [PMID: 39587036 PMCID: PMC11707835 DOI: 10.1017/erm.2024.36] [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: 12/05/2023] [Revised: 09/04/2024] [Accepted: 10/28/2024] [Indexed: 11/27/2024]
Abstract
Leishmaniasis, caused by obligate intracellular Leishmania parasites, poses a significant global health burden. The control of Leishmania infection relies on an effective T cell-dependent immune response; however, various factors impede the host’s ability to mount a successful defence. Alterations in the chemokine profile, responsible for cell trafficking to the infection site, can disrupt optimal immune responses and influence the outcome of pathogenesis by facilitating parasite persistence. This review aims to emphasize the significance of the chemokine system in T cell responses and to summarize the current knowledge on the dysregulation of chemokines and their receptors associated with different subsets of T lymphocytes during Leishmaniasis. A comprehensive understanding of the dynamic nature of the chemokine system during Leishmaniasis is crucial for the development of successful immunotherapeutic approaches.
Collapse
Affiliation(s)
- Shreya Upadhyay
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shashi Kumar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Vishal Kumar Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rahul Tiwari
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Awnish Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rajiv Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
195
|
Moore MN, Person KL, Alwin A, Krusemark C, Foster N, Ray C, Inoue A, Jackson MR, Sheedlo MJ, Barak LS, Fernandez de Velasco EM, Olson SH, Slosky LM. Design of allosteric modulators that change GPCR G protein subtype selectivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.20.624209. [PMID: 39605353 PMCID: PMC11601581 DOI: 10.1101/2024.11.20.624209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
G protein-coupled receptors (GPCRs), the largest family of drug targets, can signal through 16 subtypes of Gα proteins. Biased compounds that selectively activate therapy-relevant pathways promise to be safer, more effective medications. The determinants of bias are poorly understood, however, and rationally-designed, G protein-subtype-selective compounds are lacking. Here, using the prototypical class A GPCR neurotensin receptor 1 (NTSR1), we find that small molecules binding the intracellular GPCR-transducer interface change G protein coupling by subtype-specific and predictable mechanisms, enabling rational drug design. We demonstrate that the compound SBI-553 switches NTSR1 G protein preference by acting both as a molecular bumper and a molecular glue. Structurally, SBI-553 occludes G protein binding determinants on NTSR1, promoting association with select G protein subtypes for which an alternative, shallow-binding conformation is energetically favorable. Minor modifications to the SBI-553 scaffold produce allosteric modulators with distinct G protein subtype selectivity profiles. Selectivity profiles are probe-independent, conserved across species, and translate to differences in in vivo activity. These studies demonstrate that G protein selectivity can be tailored with small changes to a single chemical scaffold targeting the receptor-transducer interface and, as this pocket is broadly conserved, present a strategy for pathway-selective drug discovery applicable to the diverse GPCR superfamily.
Collapse
Affiliation(s)
- Madelyn N. Moore
- Department of Pharmacology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Kelsey L. Person
- Department of Pharmacology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Abigail Alwin
- Department of Pharmacology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Campbell Krusemark
- Department of Pharmacology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Noah Foster
- Department of Pharmacology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Caroline Ray
- Department of Cell Biology, Duke University, Durham, NC, USA
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Michael R. Jackson
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys, La Jolla, CA, USA
| | - Michael J. Sheedlo
- Department of Pharmacology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | | | | | - Steven H. Olson
- Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys, La Jolla, CA, USA
| | - Lauren M. Slosky
- Department of Pharmacology, University of Minnesota Twin Cities, Minneapolis, MN, USA
| |
Collapse
|
196
|
Lou F, Zhou W, Tunc-Ozdemir M, Yang J, Velazhahan V, Tate CG, Jones AM. VPS26 Moonlights as a β-Arrestin-like Adapter for a 7-Transmembrane RGS Protein in Arabidopsis thaliana. Biochemistry 2024; 63:2990-2999. [PMID: 39467170 PMCID: PMC11580166 DOI: 10.1021/acs.biochem.4c00361] [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: 06/19/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024]
Abstract
Extracellular signals perceived by 7-transmembrane (7TM)-spanning receptors initiate desensitization that involves the removal of these receptors from the plasma membrane. Agonist binding often evokes phosphorylation in the flexible C-terminal region and/or intracellular loop 3 of many 7TM G-protein-coupled receptors in animal cells, which consequently recruits a cytoplasmic intermediate adaptor, β-arrestin, resulting in clathrin-mediated endocytosis (CME) and downstream signaling such as transcriptional changes. Some 7TM receptors undergo CME without recruiting β-arrestin, but it is not clear how. Arrestins are not encoded in the Arabidopsis thaliana genome, yet Arabidopsis cells have a well-characterized signal-induced CME of a 7TM protein, designated Regulator of G Signaling 1 (AtRGS1). Here we show that a component of the retromer complex, Vacuolar Protein Sorting-Associated 26 (VPS26), binds the phosphorylated C-terminal region of AtRGS1 as a VPS26A/B heterodimer to form a complex that is required for downstream signaling. We propose that VPS26 moonlights as an arrestin-like adaptor in the CME of AtRGS1.
Collapse
Affiliation(s)
- Fei Lou
- Department
of Biology, The University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| | - Wenbin Zhou
- Department
of Biology, The University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| | - Meral Tunc-Ozdemir
- Department
of Biology, The University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| | - Jing Yang
- Department
of Biology, The University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| | - Vaithish Velazhahan
- MRC
Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, U.K.
- Gonville
and Caius College, University of Cambridge, Cambridge CB2 1TA, U.K.
| | - Christopher G. Tate
- MRC
Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, U.K.
| | - Alan M. Jones
- Department
of Biology, The University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
- Department
of Pharmacology, The University of North
Carolina at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| |
Collapse
|
197
|
Chen G, Jin Y, Chu C, Zheng Y, Yang C, Chen Y, Zhu X. A cross-tissue transcriptome-wide association study reveals GRK4 as a novel susceptibility gene for COPD. Sci Rep 2024; 14:28438. [PMID: 39558015 PMCID: PMC11574126 DOI: 10.1038/s41598-024-80122-w] [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: 08/20/2024] [Accepted: 11/15/2024] [Indexed: 11/20/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a prevalent respiratory disorder with environmental factors being the primary risk determinants. However, genetic factors also substantially contribute to the susceptibility and progression of COPD. Although genome-wide association studies (GWAS) have identified several loci associated with COPD susceptibility, the specific pathogenic genes underlying these loci, along with their biological functions and roles within regulatory networks, remain unclear. This lack of clarity constrains our ability to achieve a deeper understanding of the genetic basis of COPD. This study leveraged the FinnGen R11 genetic dataset, comprising 21,617 cases and 372,627 controls, along with GTEx V8 eQTLs data to conduct a cross-tissue transcriptome-wide association study (TWAS). Initially, we performed a cross-tissue TWAS analysis using the Unified Test for Molecular Signatures (UTMOST), followed by validation of the UTMOST findings in single tissues using the Functional Summary-based Imputation (FUSION) method and conditional and joint (COJO) analyses of the identified genes. Subsequently, candidate susceptibility genes were screened using Multi-marker Analysis of Genomic Annotation (MAGMA). The causal relationship between these candidate genes and COPD was further evaluated through summary data-based Mendelian randomization (SMR), colocalization analysis, and Mendelian randomization (MR). Additionally, the identified results were validated against the COPD dataset in the GWAS Catalog (GCST90399694). GeneMANIA was employed to further explore the functional significance of these susceptibility genes. In the cross-tissue TWAS analysis (UTMOST), we identified 17 susceptibility genes associated with COPD. Among these, a novel susceptibility gene, G protein-coupled receptor kinase 4 (GRK4), was validated through single-tissue TWAS (FUSION) and MAGMA analyses, with further confirmation via SMR, MR, and colocalization analyses. Moreover, GRK4 was validated in an independent dataset. This study identifies GRK4 as a potential novel susceptibility gene for COPD, which may influence disease risk by exacerbating inflammatory responses. The findings address gaps in previous single-tissue GWAS studies, revealing consistent expression and potential function of GRK4 across different tissues. However, considering the study's limitations, further investigation and validation of GRK4's role in COPD are warranted.
Collapse
Affiliation(s)
- Guanglei Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Yaxian Jin
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550000, Guizhou, China
| | - Cancan Chu
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Yuhao Zheng
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Changfu Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Yunzhi Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China
| | - Xing Zhu
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, Guizhou, China.
| |
Collapse
|
198
|
Crecelius JM, Manz AR, Benzow S, Marchese A. Receptor Determinants for β-Arrestin Functional Specificity at C-X-C Chemokine Receptor 5. Mol Pharmacol 2024; 106:287-297. [PMID: 39472027 PMCID: PMC11585254 DOI: 10.1124/molpharm.124.000942] [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: 05/06/2024] [Accepted: 10/07/2024] [Indexed: 11/20/2024] Open
Abstract
β-arrestins are multifaceted adaptor proteins that mediate G protein-coupled receptor (GPCR) desensitization, internalization, and signaling. It is emerging that receptor-specific determinants specify these divergent functions at GPCRs, yet this remains poorly understood. Here, we set out to identify the receptor determinants responsible for β-arrestin-mediated regulation of the chemokine receptor C-X-C motif chemokine receptor 5 (CXCR5). Using bioluminescence resonance energy transfer, we show that β-arrestin1 and β-arrestin2 are dose-dependently recruited to CXCR5 by its cognate ligand C-X-C motif chemokine ligand 13 (CXCL13). The carboxy-terminal tail of CXCR5 contains several serine/threonine residues that can be divided into three discrete phospho-site clusters based on their position relative to transmembrane domain 7. Mutagenesis experiments revealed that the distal and medial phospho-site clusters, but not the proximal, are required for agonist-stimulated β-arrestin1 or β-arrestin2 recruitment to CXCR5. Consistent with this, we provide evidence that the distal and medial, but not proximal, phospho-site clusters are required for receptor desensitization. Surprisingly, the individual phospho-site clusters are not required for agonist-stimulated internalization of CXCR5. Further, we show that CXCL13-stimulated CXCR5 internalization and ERK1/2 phosphorylation, but not desensitization, remain intact in human embryonic kidney 293 cells lacking β-arrestin1 and β-arrestin2. Our study provides evidence that β-arrestins are recruited to CXCR5 and are required for desensitization but are dispensable for internalization or signaling, suggesting that discrete receptor determinants specify the divergent functions of β-arrestins. SIGNIFICANCE STATEMENT: C-X-C motif ligand 13 (CXCL13) and C-X-C motif chemokine receptor 5 (CXCR5) are important in the immune system and are linked to diseases, yet regulation of CXCR5 signaling remains poorly understood. We provide evidence that a phospho-site cluster located at the extreme distal carboxyl-terminal tail of the receptor is responsible for β-arrestin recruitment and receptor desensitization. β-arrestins are not required for CXCL13-stimulated internalization or signaling, indicating that β-arrestins perform only one of their functions at CXCR5 and that discrete receptor determinants specify the divergent functions of β-arrestins.
Collapse
Affiliation(s)
- Joseph M Crecelius
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Aaren R Manz
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Sara Benzow
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Adriano Marchese
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
199
|
Powers AS, Khan A, Paggi JM, Latorraca NR, Souza S, Di Salvo J, Lu J, Soisson SM, Johnston JM, Weinglass AB, Dror RO. A non-canonical mechanism of GPCR activation. Nat Commun 2024; 15:9938. [PMID: 39550377 PMCID: PMC11569127 DOI: 10.1038/s41467-024-54103-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/30/2024] [Indexed: 11/18/2024] Open
Abstract
The goal of designing safer, more effective drugs has led to tremendous interest in molecular mechanisms through which ligands can precisely manipulate the signaling of G-protein-coupled receptors (GPCRs), the largest class of drug targets. Decades of research have led to the widely accepted view that all agonists-ligands that trigger GPCR activation-function by causing rearrangement of the GPCR's transmembrane helices, opening an intracellular pocket for binding of transducer proteins. Here we demonstrate that certain agonists instead trigger activation of free fatty acid receptor 1 by directly rearranging an intracellular loop that interacts with transducers. We validate the predictions of our atomic-level simulations by targeted mutagenesis; specific mutations that disrupt interactions with the intracellular loop convert these agonists into inverse agonists. Further analysis suggests that allosteric ligands could regulate the signaling of many other GPCRs via a similar mechanism, offering rich possibilities for precise control of pharmaceutically important targets.
Collapse
Affiliation(s)
- Alexander S Powers
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Aasma Khan
- Department of Quantitative Biosciences, Merck & Co., Inc., Rahway, NJ, USA
- Department of Therapeutic Proteins, Regeneron Pharmaceuticals Inc., Tarrytown, NY, USA
| | - Joseph M Paggi
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Naomi R Latorraca
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
- Biophysics Program, Stanford University, Stanford, CA, USA
- Department of Biochemistry and Molecular Biophysics, Columbia University Irving Medical Center, New York, NY, USA
| | - Sarah Souza
- Department of Quantitative Biosciences, Merck & Co., Inc., Rahway, NJ, USA
| | | | - Jun Lu
- Department of Structural Chemistry, Merck & Co., Inc., West Point, PA, USA
- Small Molecule Discovery, Zai Lab (US) LLC, Cambridge, MA, USA
| | - Stephen M Soisson
- Department of Structural Chemistry, Merck & Co., Inc., West Point, PA, USA
- Protein Therapeutics and Structural Biology, Odyssey Therapeutics, Boston, MA, USA
| | | | - Adam B Weinglass
- Department of Quantitative Biosciences, Merck & Co., Inc., Rahway, NJ, USA
| | - Ron O Dror
- Department of Computer Science, Stanford University, Stanford, CA, USA.
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA.
- Biophysics Program, Stanford University, Stanford, CA, USA.
| |
Collapse
|
200
|
Chaudhary PK, Kim S, Kunapuli SP, Kim S. Distinct Role of GRK3 in Platelet Activation by Desensitization of G Protein-Coupled Receptors. Thromb Haemost 2024. [PMID: 39419098 DOI: 10.1055/a-2442-9031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
BACKGROUND Many platelet agonists mediate their cellular effects through G protein-coupled receptors (GPCRs) to induce platelet activation, and GPCR kinases (GRKs) have been demonstrated to have crucial roles in most GPCR functions in other cell types. Here, we investigated the functional role of GRK3 and the molecular basis for the regulation of GPCR desensitization by GRK3 in platelets. METHODS We used mice lacking GRK3 as well as β-arrestin2, which has been shown to be important in GPCR function in platelets. RESULTS Platelet aggregation and dense granule secretion induced by 2-MeSADP, U46619, thrombin, and AYPGKF were significantly potentiated in both GRK3 -/- and β-arrestin2 -/- platelets compared with wild-type (WT) platelets, whereas non-GPCR agonist collagen-induced platelet aggregation and secretion were not affected. We have previously shown that GRK6 is not involved in the regulation of Gq-coupled 5HT2A and Gz-coupled α2A adrenergic receptors. Interestingly, in contrast to GRK6, platelet aggregation induced by costimulation of serotonin and epinephrine, which activate 5-HT2A and α2A adrenergic receptors, respectively, was significantly potentiated in GRK3 -/- platelets, suggesting that GRK3 is involved in general GPCR regulation. In addition, platelet aggregation in response to the second challenge of adenosine diphosphate was restored in GRK3 -/- platelets, whereas restimulation of the agonist failed to induce aggregation in WT platelets, confirming that GRK3 contributes to general GPCR desensitization. Furthermore, 2-MeSADP- and AYPGKF-induced AKT and ERK phosphorylation were significantly potentiated in GRK3 -/- platelets. Finally, GRK3 -/- mice showed shorter tail bleeding times compared with WT, indicating that GRK3 -/- mice is more susceptible to hemostasis. CONCLUSION GRK3 plays a crucial role in the regulation of platelet activation through general GPCR desensitization in platelets.
Collapse
Affiliation(s)
- Preeti K Chaudhary
- Laboratory of Veterinary Pathology and Platelet Signaling, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Sanggu Kim
- Laboratory of Veterinary Pathology and Platelet Signaling, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Satya P Kunapuli
- Sol Sherry Thrombosis Research Center and Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, United States
| | - Soochong Kim
- Laboratory of Veterinary Pathology and Platelet Signaling, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|