1
|
Sengupta P, Mukhopadhyay D. IGF1R/ARRB1 Mediated Regulation of ERK and cAMP Pathways in Response to Aβ Unfolds Novel Therapeutic Avenue in Alzheimer's Disease. Mol Neurobiol 2025; 62:8065-8083. [PMID: 39969678 DOI: 10.1007/s12035-025-04735-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 01/31/2025] [Indexed: 02/20/2025]
Abstract
IGF1R/INSR signaling is crucial for understanding Alzheimer's disease (AD) and may aid in the development of potent therapeutic strategies. This study investigated the expression and activity of these receptors and their potential to form functional hybrids in response to amyloid beta (Aβ). IGF1R, INSR, and ARRB1 were found to be upregulated in AD. The propensity for functional hybrid formation was also greater in the presence of Aβ. The association of IGF1R with ARRB1 reached a maximum at 60 min of Aβ treatment, which coincided with increased pERK activity at approximately the same time, indicating the importance of this association in pERK regulation. Knocking down IGF1R, INSR, and ARRB1 independently reduced cAMP, whereas overexpressing IGF1R significantly increased cAMP. Knocking down ARRB1 in IGF1R-overexpressing cells led to a reduction in cAMP, indicating that the interaction of ARRB1 and IGF1R possibly contributes to cAMP dysregulation. Since cAMP plays a crucial role in cognition and memory, alterations in cAMP after receptor hybridization could be significant in AD. Additionally, we noted hyperactivation of MAPK, which is associated with aberrant cellular activity, transcriptional control, and stress pathways. This finding highlights the importance of IGF1R and INSR dysregulation, which plays a major role in addition to conventional RTK signaling through multiple pathways. Here, we focused on the ARRB1 and IGF1R interaction and showed that picropodophyllin (PPP), an IGF1R-specific inhibitor, blocks this interaction and alters the ERK and cAMP status under disease conditions. Cell viability studies further revealed that the PPP substantially improved cell viability in the presence of Aβ. This highlights the role of the PPP in regulating these cascades and opens the arena for further therapeutic development for AD.
Collapse
Affiliation(s)
- Priyanka Sengupta
- Biophysical Sciences Group, 1/AF, Biddhanagar, Saha Institute of Nuclear Physics, Kolkata, 700064, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400094, India
| | - Debashis Mukhopadhyay
- Biophysical Sciences Group, 1/AF, Biddhanagar, Saha Institute of Nuclear Physics, Kolkata, 700064, India.
- Homi Bhabha National Institute, Anushaktinagar, Mumbai, 400094, India.
| |
Collapse
|
2
|
Pienkowski T, Golonko A, Bolkun L, Wawrzak-Pienkowska K, Szczerbinski L, Kretowski A, Ciborowski M, Lewandowski W, Priebe W, Swislocka R. Investigation into biased signaling, glycosylation, and drug vulnerability of acute myeloid leukemia. Pharmacol Ther 2025; 270:108848. [PMID: 40194743 DOI: 10.1016/j.pharmthera.2025.108848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/22/2025] [Accepted: 03/24/2025] [Indexed: 04/09/2025]
Abstract
Understanding and harnessing biased signaling offers significant potential for developing novel therapeutic strategies or enhancing existing treatments. By managing biased signaling, it is possible to minimize adverse effects, including toxicity, and to optimize therapeutic outcomes by selectively targeting beneficial pathways. In the context of acute myeloid leukemia (AML), a highly aggressive blood cancer characterized by the rapid proliferation of abnormal myeloid cells in the bone marrow and blood, the dysregulation of these signaling pathways, particularly those involving G protein-coupled receptors (GPCRs) and receptor tyrosine kinases (RTKs), significantly contributes to disease progression and therapeutic resistance. Traditional therapies for AML often struggle with resistance and toxicity, leading to poor patient outcomes. However, by exploiting the concept of biased signaling, researchers may be able to design drugs that selectively activate pathways that inhibit cancer cell growth while avoiding those that contribute to resistance or toxicity. Glycosylation, a key post-translational modification (PTM), plays a crucial role in biased signaling by altering receptor conformation and ligand-binding affinity, thereby affecting the outcome of biased signaling. Chemokine receptors like CXCR4, which are often overexpressed and heavily glycosylated in AML, serve as targets for therapeutic intervention. By externally inducing or inhibiting specific PTMs, it may be possible to further refine therapeutic strategies, unlocking new possibilities for developing more effective and less toxic treatments. This review highlights the importance of understanding the dynamic relationship between glycosylation and biased signaling in AML, which is essential for the development of more effective treatments and overcoming drug resistance, ultimately leading to better patient outcomes.
Collapse
Affiliation(s)
- Tomasz Pienkowski
- Clinical Research Center, Medical University of Bialystok, M. Skłodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Aleksandra Golonko
- Clinical Research Center, Medical University of Bialystok, M. Skłodowskiej-Curie 24a, 15-276 Bialystok, Poland; Department of Chemistry, Biology and Biotechnology, Bialystok University of Technology, Wiejska 45 E, 15-351 Bialystok, Poland; Waclaw Dabrowski Institute of Agricultural and Food Biotechnology State Research Institute, Rakowiecka 36, 02-532 Warsaw, Poland.
| | - Lukasz Bolkun
- Department of Hematology, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Katarzyna Wawrzak-Pienkowska
- Department of Gastroenterology, Hepatology and Internal Diseases, Voivodeship Hospital in Bialystok, 15-278 Bialystok, Poland; Department of Gastroenterology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Lukasz Szczerbinski
- Clinical Research Center, Medical University of Bialystok, M. Skłodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Adam Kretowski
- Clinical Research Center, Medical University of Bialystok, M. Skłodowskiej-Curie 24a, 15-276 Bialystok, Poland; Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Michal Ciborowski
- Clinical Research Center, Medical University of Bialystok, M. Skłodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Wlodzimierz Lewandowski
- Department of Chemistry, Biology and Biotechnology, Bialystok University of Technology, Wiejska 45 E, 15-351 Bialystok, Poland
| | - Waldemar Priebe
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1901 East Rd., Houston, TX 77054, USA
| | - Renata Swislocka
- Department of Chemistry, Biology and Biotechnology, Bialystok University of Technology, Wiejska 45 E, 15-351 Bialystok, Poland
| |
Collapse
|
3
|
Moo EV, Møller TC, Sørensen FA, Inoue A, Bräuner‐Osborne H. Arrestin-independent internalization of the GLP-1 receptor is facilitated by a GRK, clathrin, and caveolae-dependent mechanism. FEBS J 2025; 292:1675-1695. [PMID: 39756024 PMCID: PMC11970717 DOI: 10.1111/febs.17338] [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: 03/12/2024] [Revised: 07/04/2024] [Accepted: 11/19/2024] [Indexed: 01/07/2025]
Abstract
The glucagon-like peptide-1 receptor (GLP-1R) plays an important role in regulating insulin secretion and reducing body weight, making it a prominent target in the treatment of type 2 diabetes and obesity. Extensive research on GLP-1R signaling has provided insights into the connection between receptor function and physiological outcomes, such as the correlation between Gs signaling and insulin secretion, yet the exact mechanisms regulating signaling remain unclear. Here, we explore the internalization pathway of GLP-1R, which is crucial for controlling insulin release and maintaining pancreatic beta-cell function. Utilizing a reliable and sensitive time-resolved fluorescence resonance energy transfer (TR-FRET) internalization assay, combined with HEK293-derived knockout cell lines, we were able to directly compare the involvement of different endocytic machinery in GLP-1R internalization. Our findings indicate that the receptor internalizes independently of arrestin and is dependent on Gs and Gi/o activation and G protein-coupled receptor kinase phosphorylation. Mechanistically, we observed that the receptor undergoes distinct clathrin- and caveolae-mediated internalization in HEK293 cells. This study also investigated the role of arrestins in GLP-1R function and regulation. These insights into key endocytic components that are involved in the GLP-1R internalization pathway could enhance the rational design of GLP-1R therapeutics for type 2 diabetes and other GLP-1R-related diseases.
Collapse
Affiliation(s)
- Ee Von Moo
- Department of Drug Design and PharmacologyUniversity of CopenhagenDenmark
| | | | | | - Asuka Inoue
- Graduate School of Pharmaceutical SciencesTohoku UniversitySendaiJapan
- Graduate School of Pharmaceutical SciencesKyoto UniversityJapan
| | | |
Collapse
|
4
|
Bai T, He X, Liu S, He YZ, Feng J. A comprehensive review of GPR84: A novel player in pathophysiology and treatment. Int J Biol Macromol 2025; 300:140088. [PMID: 39832584 DOI: 10.1016/j.ijbiomac.2025.140088] [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: 10/28/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
G protein-coupled receptor 84 (GPR84), a member of the highly conserved rhodopsin-like superfamily, represents a promising target for therapeutic drug development. Its distinctive expression profiles in adipocytes, gut endocrine cells, and various myeloid immune cells underscore its critical roles in fundamental physiological processes, particularly in metabolic regulation and immune responses. Over the past two decades, emerging research has demonstrated that GPR84 regulates immune cell chemotaxis, phagocytosis, and inflammatory responses, playing a pivotal role in metabolic disorders, inflammatory diseases, and organ fibrosis. However, the precise molecular mechanisms by which GPR84 is involved in these diseases remain largely uncharacterized, highlighting a significant gap in our understanding. Medium-chain fatty acids (MCFAs) are considered potential endogenous ligands for GPR84. Furthermore, the development of synthetic agonists and antagonists have provided valuable pharmacological tools for analyzing the ligand-GPR84 complex structure and investigating the extensive biological functions of GPR84. Ongoing preclinical and clinical studies highlight the potential of targeting GPR84 in molecular therapies, although concerns regarding drug safety and specificity require further investigation.
Collapse
Affiliation(s)
- Tao Bai
- Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning Province, China
| | - Xin He
- Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning Province, China
| | - Shuo Liu
- Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning Province, China; The Fourth People's Hospital of Shenyang, 20 Huanghe South Street, Shenyang, Liaoning Province, China
| | - Yu-Ze He
- Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning Province, China
| | - Juan Feng
- Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning Province, China.
| |
Collapse
|
5
|
Li Y, Kawamura G, Dong Q, Li Q, Ozawa T. Near-Infrared Bioluminescence Assays for Protein-Protein Interactions and Cellular Membrane Fusion in Deep Tissues Using Split Akaluc Reconstitution. Anal Chem 2025; 97:6182-6191. [PMID: 40062634 DOI: 10.1021/acs.analchem.4c06986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Bioluminescence analysis using luciferase is an essential tool for studying biological processes in different cells. Split luciferase reconstitution is a technique that enables the analysis of biological events through the monitoring of protein-protein interactions. However, effective detection of cellular events in vivo remains challenging due to the limitation of light penetration into deep tissues and optical sensitivity. To address this, we developed a novel split luciferase reconstitution method using a near-infrared-emitting luciferase, Akaluc, and applied it to monitor two important biological events: G protein-coupled receptor (GPCR)/β-arrestin interactions and myogenic cell fusion in vivo. The developed split Akaluc reconstitution system demonstrated high sensitivity in detecting GPCR/β-arrestin interactions as well as myogenic cell fusion in vitro, enabling real-time insights into their temporal dynamics. Moreover, in vivo bioluminescence imaging successfully monitored GPCR/β-arrestin interactions in the mouse lung and the progression of myogenesis during mouse leg muscle regeneration. The split Akaluc reconstitution method will be a versatile tool for both in vitro and in vivo analyses of protein-protein interactions and cell fusion events. This system holds significant potential for advancing drug development, especially in the screening of GPCR-targeted therapeutic and myogenesis-promoting compounds in animal models.
Collapse
Affiliation(s)
- Yiling Li
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Genki Kawamura
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Qi Dong
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Qiaojing Li
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takeaki Ozawa
- Department of Chemistry, School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
6
|
Patnaik R, Varghese RL, Banerjee Y. Selective Modulation of PAR-2-Driven Inflammatory Pathways by Oleocanthal: Attenuation of TNF-α and Calcium Dysregulation in Colorectal Cancer Models. Int J Mol Sci 2025; 26:2934. [PMID: 40243559 PMCID: PMC11988659 DOI: 10.3390/ijms26072934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 03/15/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Colorectal cancer (CRC) remains a principal contributor to oncological mortality worldwide, with chronic inflammation serving as a fundamental driver of its pathogenesis. Protease-activated receptor-2 (PAR-2), a G-protein-coupled receptor, orchestrates inflammation-driven tumorigenesis by potentiating NF-κB and Wnt/β-catenin signaling, thereby fostering epithelial-mesenchymal transition (EMT), immune evasion, and therapeutic resistance. Despite its pathological significance, targeted modulation of PAR-2 remains an underexplored avenue in CRC therapeutics. Oleocanthal (OC), a phenolic constituent of extra virgin olive oil, is recognized for its potent anti-inflammatory and anti-cancer properties; however, its regulatory influence on PAR-2 signaling in CRC is yet to be elucidated. This study interrogates the impact of OC on PAR-2-mediated inflammatory cascades using HT-29 and Caco-2 CRC cell lines subjected to lipopolysaccharide (LPS)-induced activation of PAR-2. Expression levels of PAR-2 and TNF-α were quantified through Western blotting and RT-PCR, while ELISA assessed TNF-α secretion. Intracellular calcium flux, a pivotal modulator of PAR-2-driven oncogenic inflammation, was evaluated via Fluo-4 calcium assays. LPS markedly elevated PAR-2 expression at both mRNA and protein levels in CRC cells (p < 0.01, one-way ANOVA). OC administration (20-150 μg/mL) elicited a dose-dependent suppression of PAR-2, with maximal inhibition at 100-150 μg/mL (p < 0.001, Tukey's post hoc test). Concomitant reductions in TNF-α transcription (p < 0.01) and secretion (p < 0.001) were observed, corroborating the anti-inflammatory efficacy of OC. Additionally, OC ameliorated LPS-induced calcium dysregulation, restoring intracellular calcium homeostasis in a concentration-dependent manner (p < 0.01). Crucially, OC exhibited selectivity for PAR-2, leaving PAR-1 expression unaltered (p > 0.05), underscoring its precision as a therapeutic agent. These findings position OC as a selective modulator of PAR-2-driven inflammation in CRC, disrupting the pro-tumorigenic microenvironment through attenuation of TNF-α secretion, calcium dysregulation, and oncogenic signaling pathways. This study furnishes mechanistic insights into OC's potential as a nutraceutical intervention in inflammation-associated CRC. Given the variability in OC bioavailability and content in commercial olive oil, future investigations should delineate optimal dosing strategies and in vivo efficacy to advance its translational potential in CRC therapy.
Collapse
Affiliation(s)
- Rajashree Patnaik
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai 505055, United Arab Emirates; (R.P.); (R.L.V.)
| | - Riah Lee Varghese
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai 505055, United Arab Emirates; (R.P.); (R.L.V.)
| | - Yajnavalka Banerjee
- Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Health, Dubai 505055, United Arab Emirates; (R.P.); (R.L.V.)
- Centre for Medical Education, School of Medicine, University of Dundee Ninewells Hospital Dundee, Dundee DD2 1SG, UK
| |
Collapse
|
7
|
Park JS, Ma YQ, Wang F, Ma H, Sui G, Rustamov N, Han M, Son Y, Park CW, Han SB, Hong JT, Jeong LS, Lee J, Roh YS. A3AR antagonism mitigates metabolic dysfunction-associated steatotic liver disease by exploiting monocyte-derived Kupffer cell necroptosis and inflammation resolution. Metabolism 2025; 164:156114. [PMID: 39732364 DOI: 10.1016/j.metabol.2024.156114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND & AIMS Metabolic dysfunction-associated steatotic liver (MASLD) progression is driven by chronic inflammation and fibrosis, largely influenced by Kupffer cell (KC) dynamics, particularly replenishment of pro-inflammatory monocyte-derived KCs (MoKCs) due to increased death of embryo-derived KCs. Adenosine A3 receptor (A3AR) plays a key role in regulating metabolism and immune responses, making it a promising therapeutic target. This study aimed to investigate the impact of selective A3AR antagonism for regulation of replenished MoKCs, thereby improving MASLD. APPROACH & RESULTS A3AR expression was significantly elevated in KCs from both patients with MASLD and fast-food diet (FFD)-fed mice. A3AR knockout (KO) mice displayed marked improvements in hepatic inflammation and fibrosis along with a reduction in CLEC4F-positive KCs. The spatial transcriptomics of these KCs revealed disrupted mitochondrial integrity, increased oxidative stress, and enhanced cell death due to A3AR deletion. Similarly, in vivo FM101 treatment, a highly potent and selective antagonist of A3AR with a truncated 4'-thioadenosine structure, mitigated FFD-induced MASLD in mice. Mechanistically, FM101 induces β-arrestin2-mediated A3AR degradation, leading to mitochondrial dysfunction-mediated necroptosis in KCs. Consistently, A3AR was highly expressed in monocyte-derived macrophages in MASLD patients, with strong correlations with macrophage activation and monocyte chemoattractant gene sets. Thus, FM101 induced necroptosis in pro-inflammatory MoKCs, facilitating anti-inflammatory effects. CONCLUSIONS This study demonstrated that inhibiting A3AR via FM101 or genetic deletion alleviates MASLD by inducing mitochondrial dysfunction and subsequent necroptosis in MoKCs, establishing FM101 as a promising therapeutic strategy for MASLD.
Collapse
Affiliation(s)
- Jeong-Su Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Yuan-Qiang Ma
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Feng Wang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Hwan Ma
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Guoyan Sui
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Nodir Rustamov
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Minyeong Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Yejin Son
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Chun-Woong Park
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea
| | - Lak Shin Jeong
- Research and Development Center, Future Medicine Co., Ltd, Seongnam, South Korea; College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Jin Lee
- Department of Pathology, School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Yoon Seok Roh
- College of Pharmacy and Medical Research Center, Chungbuk National University, Cheongju, Chungbuk, South Korea.
| |
Collapse
|
8
|
Ju J, Li Z, Liu J, Peng X, Gao F. Biased Opioid Receptor Agonists: Balancing Analgesic Efficacy and Side-Effect Profiles. Int J Mol Sci 2025; 26:1862. [PMID: 40076488 PMCID: PMC11899445 DOI: 10.3390/ijms26051862] [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: 12/19/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 03/14/2025] Open
Abstract
Opioids are the most effective option for severe pain. However, it is well documented that the side effects associated with prolonged opioid use significantly constrain dosage in the clinical setting. Recently, researchers have concentrated on the development of biased opioid receptor agonists that preferentially activate the G protein signaling pathway over β-arrestin signaling. This approach is based on the hypothesis that G protein signaling mediates analgesic effects, whereas β-arrestin signaling is implicated in adverse side effects. Although certain studies have demonstrated that the absence or inhibition of β-arrestin signaling can mitigate the incidence of side effects, recent research appears to challenge these earlier findings. In-depth investigations into biased signal transduction of opioid receptor agonists have been conducted, potentially offering novel insights for the development of biased opioid receptors. Consequently, this review elucidates the contradictory roles of β-arrestin signaling in the adverse reactions associated with opioid receptor activation. Furthermore, a comparative analysis was conducted to evaluate the efficacy of the classic G protein-biased agonists, TRV130 and PZM21, relative to the traditional non-biased agonist morphine. This review aims to inform the development of novel analgesic drugs that can optimize therapeutic efficacy and safety, while minimizing adverse reactions to the greatest extent possible.
Collapse
Affiliation(s)
| | | | | | | | - Feng Gao
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (J.J.); (Z.L.); (J.L.); (X.P.)
| |
Collapse
|
9
|
Teixeira LB, Blouin MJ, Le Gouill C, Picard LP, Costa-Neto CM, Bouvier M, Parreiras-E-Silva LT. Sustained Gα s signaling mediated by vasopressin type 2 receptors is ligand dependent but endocytosis and β-arrestin independent. Sci Signal 2025; 18:eadf6206. [PMID: 40136046 DOI: 10.1126/scisignal.adf6206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 07/01/2024] [Accepted: 01/29/2025] [Indexed: 03/27/2025]
Abstract
The canonical model of G protein-coupled receptor (GPCR) signaling comprises G protein activation at the plasma membrane, followed by receptor phosphorylation and β-arrestin recruitment, which leads to receptor desensitization and endocytosis. However, the activation of some GPCRs results in sustained G protein signaling from intracellular compartments in a manner reportedly dependent on β-arrestin and receptor endocytosis. The vasopressin type 2 receptor (V2R) can be activated by two structurally similar hormones, arginine vasopressin and oxytocin, both of which stimulate the production of the second messenger cyclic adenosine monophosphate (cAMP). In this study, we showed that sustained V2R signaling and endosomal Gαs (stimulatory G protein alpha subunit) translocation could occur without β-arrestin-mediated receptor endocytosis and was primarily controlled by the residence time of the ligand on the receptor. β-Arrestin had opposing effects on sustained signaling: It facilitated receptor internalization into endosomes, where it activated Gαs, and promoted cAMP production from this compartment. However, β-arrestin-mediated receptor endocytosis also induced ligand dissociation due to the acidic endosomal environment, thereby limiting the signal. Overall, our data suggest that signals originating at the plasma membrane play a dominant role in sustained V2R signaling stimulated by arginine vasopressin.
Collapse
Affiliation(s)
- Larissa B Teixeira
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC H3T 1J4, Canada
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP 14.049-900, Brazil
| | - Marie-José Blouin
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Christian Le Gouill
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Louis-Philippe Picard
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Claudio M Costa-Neto
- Department of Radiology and Oncology, Faculty of Medicine, University of São Paulo, São Paulo, SP 01246-903, Brazil
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Lucas T Parreiras-E-Silva
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montreal, QC H3T 1J4, Canada
- Department of BioMolecular Sciences, Ribeirão Preto Pharmaceutical Sciences School, University of São Paulo, Ribeirão Preto, SP 14.040-903, Brazil
| |
Collapse
|
10
|
Saca VR, Huber T, Sakmar TP. G protein-coupled receptor-targeted proteolysis-targeting chimeras in cancer therapeutics. Mol Pharmacol 2025; 107:100013. [PMID: 40023512 DOI: 10.1016/j.molpha.2024.100013] [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: 10/07/2024] [Accepted: 12/05/2024] [Indexed: 03/04/2025] Open
Abstract
G protein-coupled receptors (GPCRs) comprise a family of heptahelical membrane proteins that mediate intracellular and intercellular transmembrane signaling. Defects in GPCR signaling pathways are implicated in the pathophysiology of many diseases, including cardiovascular disease, endocrinopathies, immune disorders, and cancer. Although GPCRs are attractive drug targets, only a small number of Food and Drug Administration-approved anticancer therapeutics target GPCRs. Targeted protein degradation (TPD) technology allows for the direct modulation of the cellular expression level of a protein of interest. TPD methods such as proteolysis-targeting chimeras (PROTACs) use the ubiquitin-proteasome system to degrade a protein of interest selectively. Although the PROTAC system has not been widely applied to GPCRs and other membrane proteins, there is evidence that PROTACs or other TPD methods could be applied to the GPCRome. Current GPCR PROTACs show the feasibility of using PROTACs to degrade GPCRs; however, the degradation mechanism for some of these GPCR PROTACs is uncertain. Additional studies aimed at elucidating the degradation mechanism of GPCRs with PROTACs are necessary. Discovery of new allosteric intracellular small molecule binders of GPCRs will be required for the development of intracellularly oriented PROTACs. Promising early results in targeted degradation of GPCRs suggest that TPD drug discovery platforms will be useful in developing PROTACs targeting pathological GPCRs. SIGNIFICANCE STATEMENT: Aberrant signaling of G protein-coupled receptors (GPCRs) can contribute to the pathophysiology of cancer. Although GPCRs are generally highly attractive drug targets, many individual GPCRs are currently undrugged using traditional drug discovery approaches. Targeted protein degradation technologies, such as proteolysis-targeting chimeras, provide a new approach to drug discovery for targeting previously undruggable GPCRs relevant to the molecular pathophysiology of cancer.
Collapse
Affiliation(s)
- Victoria R Saca
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York; Tri-Institutional PhD Program in Chemical Biology, New York, New York
| | - Thomas Huber
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York
| | - Thomas P Sakmar
- Laboratory of Chemical Biology and Signal Transduction, The Rockefeller University, New York, New York.
| |
Collapse
|
11
|
Carino CMC, Hiratsuka S, Kise R, Nakamura G, Kawakami K, Yanagawa M, Inoue A. Signal profiles and spatial regulation of β-arrestin recruitment through Gβ 5 and GRK3 at the μ-opioid receptor. Eur J Pharmacol 2025; 987:177151. [PMID: 39579957 DOI: 10.1016/j.ejphar.2024.177151] [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: 06/21/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 11/25/2024]
Abstract
The μ-opioid receptor (MOR) is a G-protein-coupled receptor (GPCR) that mediates both analgesic effects and adverse effects of opioid drugs. Despite extensive efforts to develop a signal-biased drug, drugs with sufficiently reduced side effects have not been established, in part owing to lack of comprehensive signal transducer profiles of MOR. In this study, by profiling the activity of signal transducers including G proteins and GPCR kinases (GRKs), we revealed an unprecedented mechanism of selective GRK3 activation by Gβ5, leading to β-arrestin recruitment. By utilizing multiple genome-edited cell lines and functional assays, we found that oliceridine, an FDA-approved G-protein-biased agonist, selectively activates Gαz- and GRK3-mediated signaling. Notably, among the five Gβ subtypes, only Gβ5 distinguishes GRK3 from GRK2. Using single-molecule imaging, we found that GRK3 is recruited to the plasma membrane upon MOR agonist stimulation by Gβ1 and Gβ5, yet their interaction dynamics with GRK3 and mechanisms of action are different. Furthermore, particle diffusion analysis suggests that Gβ5 is enriched in confined membrane domains, through which GRK3 is recruited to the plasma membrane in a freely diffusible state, thereby allowing GRK3 to efficiently interact with MOR. These findings provide a mechanism by which MOR agonists rely on a specific Gα-Gβ-GRK axis to induce β-arrestin recruitment.
Collapse
Affiliation(s)
- Carlo Marion C Carino
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Suzune Hiratsuka
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Ryoji Kise
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan.
| | - Gaku Nakamura
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Kouki Kawakami
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Masataka Yanagawa
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan; Cellular Informatics Laboratory, RIKEN Cluster for Pioneering Research, Wako, Saitama, 351-0198, Japan
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan; Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan.
| |
Collapse
|
12
|
Boon K, Vanalken N, Szpakowska M, Chevigné A, Schols D, Van Loy T. High-affinity ELR+ chemokine ligands show G protein bias over β-arrestin recruitment and receptor internalization in CXCR1 signaling. J Biol Chem 2025; 301:108044. [PMID: 39615686 PMCID: PMC11732455 DOI: 10.1016/j.jbc.2024.108044] [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: 06/26/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024] Open
Abstract
The human CXC chemokine receptor 1 (CXCR1), a G protein-coupled receptor (GPCR), plays significant roles in inflammatory diseases and cancer. While CXCL8 is a well-established high-affinity ligand for CXCR1, there is no consensus regarding the binding ability of the other ELR+ chemokines (CXCL1-3 and CXCL5-8). Since research has predominantly focused on CXCL8-mediated CXCR1 signaling, insight into potential signaling bias induced by different CXCR1 ligands is lacking. Therefore, in this study we first compared and clarified the binding ability of all ELR+ chemokines using a competition binding assay. In this assay CXCL1-3 and CXCL5 behaved as low-affinity ligands while CXCL6-8 were high affinity ligands. We further investigated potential ligand bias within the CXCR1 signaling system. Using NanoBRET-based assays heterotrimeric G protein dissociation, β-arrestin recruitment and receptor internalization induced by chemokine binding to CXCR1 were investigated. A quantitative and qualitative investigation of ligand bias showed that the high-affinity ELR+ chemokines were biased towards G protein activation over β-arrestin recruitment and receptor internalization, when CXCL8 was used as a reference ligand.
Collapse
Affiliation(s)
- Katrijn Boon
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular Structural and Translational Virology Research Group, Leuven, Belgium
| | - Nathan Vanalken
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular Structural and Translational Virology Research Group, Leuven, Belgium
| | - Martyna Szpakowska
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Andy Chevigné
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| | - Dominique Schols
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular Structural and Translational Virology Research Group, Leuven, Belgium
| | - Tom Van Loy
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Molecular Structural and Translational Virology Research Group, Leuven, Belgium.
| |
Collapse
|
13
|
Gómez-Cañas M, Rodríguez-Cueto C, Fernández-Ruiz J, de Lago E. Development of Novel Neuroprotective Agents Targeting GPCR Using in Vitro Assays. Methods Mol Biol 2025; 2899:339-366. [PMID: 40067634 DOI: 10.1007/978-1-0716-4386-0_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
This chapter aims to elucidate an example of the drug development process for a G protein-coupled receptor (GPCR), focusing specifically on the cannabinoid receptors CB1 and CB2. Various techniques can be employed for different purposes when characterizing a new compound targeting these receptors.Initially, the compound affinity for both cannabinoid receptors is assessed through binding competition assays. Subsequently, the compound's functionality on cannabinoid receptors at intracellular signaling levels is investigated. Cannabinoid receptors predominantly interact with G proteins, although other interactors such as β-arrestins, SGIP1, CRIP1a/b, BiP, and GAP43 have been identified. So, selective functionality or biased agonism, favoring one signaling pathway over another, has been a focus of recent research efforts. The ability of compounds to activate the α subunit of G-proteins is assessed through a [35S]-GTPγS binding assays.With the above experimental tools, it is possible to investigate cannabinoids in relation to their interaction with cannabinoid receptors, which may be useful in understanding their pharmacological effects on numerous biological processes. Of high interest is the role of these receptors in the control of cell integrity and survival, which explains the study of cannabinoids and, in general, of the modulation of the endocannabinoid system (ECS) as a therapeutic strategy on neurodegenerative diseases due to its involvement in mitigating neuroinflammation, excitotoxicity, and oxidative stress. In vitro models employing cell lines and toxin mimics of neurodegenerative disease pathology are utilized to evaluate the compounds' potential therapeutic effects.In summary, the drug discovery process for GPCRs involves a comprehensive assessment of compound affinity, functionality, and physiological effects, ultimately aiming to identify novel therapeutic strategies for various human disorders.
Collapse
Affiliation(s)
- María Gómez-Cañas
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Carmen Rodríguez-Cueto
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Javier Fernández-Ruiz
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Eva de Lago
- Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.
| |
Collapse
|
14
|
Pacheco J, Peña KA, Savransky S, Gidon A, Hammond GRV, Janetzko J, Vilardaga JP. Fast-diffusing receptor collisions with slow-diffusing peptide ligand assemble the ternary parathyroid hormone-GPCR-arrestin complex. Nat Commun 2024; 15:10499. [PMID: 39627206 PMCID: PMC11615292 DOI: 10.1038/s41467-024-54772-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 11/19/2024] [Indexed: 12/06/2024] Open
Abstract
The assembly of a peptide ligand, its receptor, and β-arrestin (βarr) into a ternary complex within the cell membrane is a crucial aspect of G protein-coupled receptor (GPCR) signaling. We explore this assembly by attaching fluorescent moieties to the parathyroid hormone (PTH) type 1 receptor (PTH1R), using PTH as a prototypical peptide hormone, along with βarr and clathrin, and recording dual-color single-molecule imaging at the plasma membrane of live cells. Here we show that PTH1R exhibits a near-Brownian diffusion, whereas unbound hormone displays limited mobility and slow lateral diffusion at the cell surface. The formation of the PTH-PTH1R-βarr complex occurs in three sequential steps: (1) receptor and ligand collisions, (2) phosphoinositide (PIP3)-dependent recruitment and conformational change of βarr molecules at the plasma membrane, and (3) collision of most βarr molecules with the ligand-bound receptor within clathrin clusters. Our results elucidate the non-random pathway by which PTH-PTH1R-βarr complex is formed and unveil the critical role of PIP3 in regulating GPCR signaling.
Collapse
Affiliation(s)
- Jonathan Pacheco
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Karina A Peña
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Sofya Savransky
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
- Graduate Program in Molecular Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Alexandre Gidon
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
- Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Gerald R V Hammond
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - John Janetzko
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Jean-Pierre Vilardaga
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA.
- Graduate Program in Molecular Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA.
- U.S. Department of Veterans Affairs, Pittsburgh Healthcare System, Pittsburgh, PA, 15240, USA.
| |
Collapse
|
15
|
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
|
16
|
Weng L, Hong H, Zhang Q, Xiao C, Zhang Q, Wang Q, Huang J, Lai D. Sleep Deprivation Triggers the Excessive Activation of Ovarian Primordial Follicles via β2 Adrenergic Receptor Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402393. [PMID: 39229959 PMCID: PMC11538700 DOI: 10.1002/advs.202402393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/23/2024] [Indexed: 09/05/2024]
Abstract
Sleep deprivation (SD) is observed to adversely affect the reproductive health of women. However, its precise physiological mechanisms remain largely elusive. In this study, using a mouse model of SD, it is demonstrated that SD induces the depletion of ovarian primordial follicles, a phenomenon not attributed to immune-mediated attacks or sympathetic nervous system activation. Rather, the excessive secretion of stress hormones, namely norepinephrine (NE) and epinephrine (E), by overactive adrenal glands, has emerged as a key mediator. The communication pathway mediated by the KIT ligand (KITL)-KIT between granulosa cells and oocytes plays a pivotal role in primordial follicle activation. SD heightened the levels of NE/E that stimulates the activation of the KITL-KIT/PI3K and mTOR signaling cascade in an β2 adrenergic receptor (ADRB2)-dependent manner, thereby promoting primordial follicle activation and consequent primordial follicle loss in vivo. In vitro experiments further corroborate these observations, revealing that ADRB2 upregulates KITL expression in granulosa cells via the activation of the downstream cAMP/PKA pathway. Together, these results reveal the significant involvement of ADRB2 signaling in the depletion of ovarian primordial follicles under sleep-deprived conditions. Additionally, ADRB2 antagonists are proposed for the treatment or prevention of excessive activation of primordial follicles induced by SD.
Collapse
Affiliation(s)
- Lichun Weng
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Hanqing Hong
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Qinyu Zhang
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Chengqi Xiao
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Qiuwan Zhang
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Qian Wang
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| | - Ju Huang
- Songjiang Hospital and Songjiang Research InstituteShanghai Key Laboratory of Emotions and Affective DisordersShanghai Jiao Tong University School of MedicineShanghai201600China
| | - Dongmei Lai
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200030China
- Shanghai Key Laboratory of Embryo Original DiseasesShanghai200030China
| |
Collapse
|
17
|
Manti S, Gambadauro A, Galletta F, Ruggeri P, Piedimonte G. Update on the Role of β2AR and TRPV1 in Respiratory Diseases. Int J Mol Sci 2024; 25:10234. [PMID: 39408565 PMCID: PMC11477158 DOI: 10.3390/ijms251910234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Respiratory diseases (RDs) constitute a common public health problem both in industrialized and developing countries. The comprehension of the pathophysiological mechanisms underlying these conditions and the development of new therapeutic strategies are critical for improving the quality of life of affected patients. β2-adrenergic receptor (β2AR) and transient receptor potential vanilloid 1 (TRPV1) are both involved in physiological responses in the airways. β2AR is implicated in bronchodilation, mucociliary clearance, and anti-inflammatory effects, while TRPV1 is involved in the mediation of pain and cough reflexes. In RDs, such as respiratory infections, asthma, chronic obstructive pulmonary disease (COPD), and cystic fibrosis, the concentration and expression of these receptors can be altered, leading to significant consequences. In this review, we provided an update on the literature about the role of β2AR and TRPV1 in these conditions. We reported how the diminished or defective expression of β2AR during viral infections or prolonged therapy with β2-agonists can increase the severity of these pathologies and impact the prognosis. Conversely, the role of TRPV1 was pivotal in neuroinflammation, and its modulation could lead to innovative treatment strategies in specific patients. We indicate future perspectives and potential personalized treatments in RDs through a comprehensive analysis of the roles of these receptors in the physiological and pathological mechanisms of these pathologies.
Collapse
Affiliation(s)
- Sara Manti
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, Via Consolare Valeria 1, 98124 Messina, Italy; (S.M.); (F.G.)
| | - Antonella Gambadauro
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, Via Consolare Valeria 1, 98124 Messina, Italy; (S.M.); (F.G.)
| | - Francesca Galletta
- Pediatric Unit, Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, University of Messina, Via Consolare Valeria 1, 98124 Messina, Italy; (S.M.); (F.G.)
| | - Paolo Ruggeri
- Pulmonology Unit, Department of Biomedical and Dental Sciences, University of Messina, Via Consolare Valeria 1, 98124 Messina, Italy
| | - Giovanni Piedimonte
- Office for Research and Departments of Pediatrics, Biochemistry, and Molecular Biology, Tulane University, New Orleans, LA 70112, USA;
| |
Collapse
|
18
|
Zhao M, Cao N, Gu H, Xu J, Xu W, Zhang D, Wei TYW, Wang K, Guo R, Cui H, Wang X, Guo X, Li Z, He K, Li Z, Zhang Y, Shyy JYJ, Dong E, Xiao H. AMPK Attenuation of β-Adrenergic Receptor-Induced Cardiac Injury via Phosphorylation of β-Arrestin-1-ser330. Circ Res 2024; 135:651-667. [PMID: 39082138 DOI: 10.1161/circresaha.124.324762] [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: 05/21/2024] [Revised: 07/16/2024] [Accepted: 07/22/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND β-adrenergic receptor (β-AR) overactivation is a major pathological cue associated with cardiac injury and diseases. AMPK (AMP-activated protein kinase), a conserved energy sensor, regulates energy metabolism and is cardioprotective. However, whether AMPK exerts cardioprotective effects via regulating the signaling pathway downstream of β-AR remains unclear. METHODS Using immunoprecipitation, mass spectrometry, site-specific mutation, in vitro kinase assay, and in vivo animal studies, we determined whether AMPK phosphorylates β-arrestin-1 at serine (Ser) 330. Wild-type mice and mice with site-specific mutagenesis (S330A knock-in [KI]/S330D KI) were subcutaneously injected with the β-AR agonist isoproterenol (5 mg/kg) to evaluate the causality between β-adrenergic insult and β-arrestin-1 Ser330 phosphorylation. Cardiac transcriptomics was used to identify changes in gene expression from β-arrestin-1-S330A/S330D mutation and β-adrenergic insult. RESULTS Metformin could decrease cAMP/PKA (protein kinase A) signaling induced by isoproterenol. AMPK bound to β-arrestin-1 and phosphorylated Ser330 with the highest phosphorylated mass spectrometry score. AMPK activation promoted β-arrestin-1 Ser330 phosphorylation in vitro and in vivo. Neonatal mouse cardiomyocytes overexpressing β-arrestin-1-S330D (active form) inhibited the β-AR/cAMP/PKA axis by increasing PDE (phosphodiesterase) 4 expression and activity. Cardiac transcriptomics revealed that the differentially expressed genes between isoproterenol-treated S330A KI and S330D KI mice were mainly involved in immune processes and inflammatory response. β-arrestin-1 Ser330 phosphorylation inhibited isoproterenol-induced reactive oxygen species production and NLRP3 (NOD-like receptor protein 3) inflammasome activation in neonatal mouse cardiomyocytes. In S330D KI mice, the β-AR-activated cAMP/PKA pathways were attenuated, leading to repressed inflammasome activation, reduced expression of proinflammatory cytokines, and mitigated macrophage infiltration. Compared with S330A KI mice, S330D KI mice showed diminished cardiac fibrosis and improved cardiac function upon isoproterenol exposure. However, the cardiac protection exerted by AMPK was abolished in S330A KI mice. CONCLUSIONS AMPK phosphorylation of β-arrestin-1 Ser330 potentiated PDE4 expression and activity, thereby inhibiting β-AR/cAMP/PKA activation. Subsequently, β-arrestin-1 Ser330 phosphorylation blocks β-AR-induced cardiac inflammasome activation and remodeling.
Collapse
Affiliation(s)
- Mingming Zhao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Ning Cao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital (N.C.), Capital Medical University, Beijing, China
| | - Huijun Gu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Jiachao Xu
- Laboratory for Clinical Medicine (N.C.), Capital Medical University, Beijing, China
| | - Wenli Xu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China (W.X., E.D., H.X.)
| | - Di Zhang
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies (D.Z., Zhiyuan Li), Peking University, Beijing, China
| | - Tong-You Wade Wei
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-Y.W.W., J.Y.-J.S.)
| | - Kang Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Ruiping Guo
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Hongtu Cui
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Xiaofeng Wang
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (X.W.)
| | - Xin Guo
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Zhiyuan Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies (D.Z., Zhiyuan Li), Peking University, Beijing, China
| | - Kangmin He
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China (J.X., K.H.)
- University of Chinese Academy of Sciences, Beijing, China (K.H.)
| | - Zijian Li
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Youyi Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - John Y-J Shyy
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-Y.W.W., J.Y.-J.S.)
| | - Erdan Dong
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- Institute of Cardiovascular Sciences (E.D.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China (W.X., E.D., H.X.)
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China (W.X., E.D., H.X.)
| |
Collapse
|
19
|
Kukkonen JP, Jacobson LH, Hoyer D, Rinne MK, Borgland SL. International Union of Basic and Clinical Pharmacology CXIV: Orexin Receptor Function, Nomenclature and Pharmacology. Pharmacol Rev 2024; 76:625-688. [PMID: 38902035 DOI: 10.1124/pharmrev.123.000953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/02/2024] [Accepted: 06/06/2024] [Indexed: 06/22/2024] Open
Abstract
The orexin system consists of the peptide transmitters orexin-A and -B and the G protein-coupled orexin receptors OX1 and OX2 Orexin receptors are capable of coupling to all four families of heterotrimeric G proteins, and there are also other complex features of the orexin receptor signaling. The system was discovered 25 years ago and was immediately identified as a central regulator of sleep and wakefulness; this is exemplified by the symptomatology of the disorder narcolepsy with cataplexy, in which orexinergic neurons degenerate. Subsequent translation of these findings into drug discovery and development has resulted to date in three clinically used orexin receptor antagonists to treat insomnia. In addition to sleep and wakefulness, the orexin system appears to be a central player at least in addiction and reward, and has a role in depression, anxiety and pain gating. Additional antagonists and agonists are in development to treat, for instance, insomnia, narcolepsy with or without cataplexy and other disorders with excessive daytime sleepiness, depression with insomnia, anxiety, schizophrenia, as well as eating and substance use disorders. The orexin system has thus proved an important regulator of numerous neural functions and a valuable drug target. Orexin prepro-peptide and orexin receptors are also expressed outside the central nervous system, but their potential physiological roles there remain unknown. SIGNIFICANCE STATEMENT: The orexin system was discovered 25 years ago and immediately emerged as an essential sleep-wakefulness regulator. This discovery has tremendously increased the understanding of these processes and has thus far resulted in the market approval of three orexin receptor antagonists, which promote more physiological aspects of sleep than previous hypnotics. Further, orexin receptor agonists and antagonists with different pharmacodynamic properties are in development since research has revealed additional potential therapeutic indications. Orexin receptor signaling is complex and may represent novel features.
Collapse
Affiliation(s)
- Jyrki P Kukkonen
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Laura H Jacobson
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Daniel Hoyer
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Maiju K Rinne
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| | - Stephanie L Borgland
- Department of Pharmacology, Medicum, University of Helsinki, Helsinki, Finland (J.P.K., M.K.R.); Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne (D.H., L.H.J.), The Florey (D.H., L.H.J.), Parkville, Victoria, Australia; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California (D.H.); and Department of Physiology and Pharmacology, University of Calgary, Calgary Canada (S.L.B.)
| |
Collapse
|
20
|
Lattanzi R, Casella I, Fullone MR, Vincenzi M, Maftei D, Miele R. Mapping the interaction site for β-arrestin-2 in the prokineticin 2 receptor. Cell Signal 2024; 119:111175. [PMID: 38631405 DOI: 10.1016/j.cellsig.2024.111175] [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: 01/26/2024] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
G protein-coupled receptors (GPCRs) are a family of cell membrane receptors that couple and activate heterotrimeric G proteins and their associated intracellular signalling processes after ligand binding. Although the carboxyl terminal of the receptors is essential for this action, it can also serve as a docking site for regulatory proteins such as the β-arrestins. Prokineticin receptors (PKR1 and PKR2) are a new class of GPCRs that are able to activate different classes of G proteins and form complexes with β-arrestins after activation by the endogenous agonists PK2. The aim of this work was to define the molecular determinants within PKR2 that are required for β-arrestin-2 binding and to investigate the role of β-arrestin-2 in the signalling pathways induced by PKR2 activation. Our data show that PKR2 binds constitutively to β-arrestin-2 and that this process occurs through the core region of the receptor without being affected by the carboxy-terminal region. Indeed, a PKR2 mutant lacking the carboxy-terminal amino acids retains the ability to bind constitutively to β-arrestin-2, whereas a mutant lacking the third intracellular loop does not. Overall, our data suggest that the C-terminus of PKR2 is critical for the stability of the β-arrestin-2-receptor complex in the presence of PK2 ligand. This leads to the β-arrestin-2 conformational change required to initiate intracellular signalling that ultimately leads to ERK phosphorylation and activation.
Collapse
Affiliation(s)
- R Lattanzi
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - I Casella
- Dipartimento del Farmaco, Istituto Superiore di Sanita, I-00161 Rome, Italy
| | - M R Fullone
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - M Vincenzi
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - D Maftei
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - R Miele
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy.
| |
Collapse
|
21
|
Solís KH, Romero-Ávila MT, Rincón-Heredia R, García-Sáinz JA. Lysophosphatidic Acid Receptor 3 (LPA3): Signaling and Phosphorylation Sites. Int J Mol Sci 2024; 25:6491. [PMID: 38928196 PMCID: PMC11203643 DOI: 10.3390/ijms25126491] [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: 04/05/2024] [Revised: 06/03/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
LPA3 receptors were expressed in TREx HEK 293 cells, and their signaling and phosphorylation were studied. The agonist, lysophosphatidic acid (LPA), increased intracellular calcium and ERK phosphorylation through pertussis toxin-insensitive processes. Phorbol myristate acetate, but not LPA, desensitizes LPA3-mediated calcium signaling, the agonists, and the phorbol ester-induced LPA3 internalization. Pitstop 2 (clathrin heavy chain inhibitor) markedly reduced LPA-induced receptor internalization; in contrast, phorbol ester-induced internalization was only delayed. LPA induced rapid β-arrestin-LPA3 receptor association. The agonist and the phorbol ester-induced marked LPA3 receptor phosphorylation, and phosphorylation sites were detected using mass spectrometry. Phosphorylated residues were detected in the intracellular loop 3 (S221, T224, S225, and S229) and in the carboxyl terminus (S321, S325, S331, T333, S335, Y337, and S343). Interestingly, phosphorylation sites are within sequences predicted to constitute β-arrestin binding sites. These data provide insight into LPA3 receptor signaling and regulation.
Collapse
Affiliation(s)
- K. Helivier Solís
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México 04510, Mexico; (K.H.S.); (M.T.R.-Á.)
| | - M. Teresa Romero-Ávila
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México 04510, Mexico; (K.H.S.); (M.T.R.-Á.)
| | - Ruth Rincón-Heredia
- Unidad de Imagenología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México 04510, Mexico;
| | - J. Adolfo García-Sáinz
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México 04510, Mexico; (K.H.S.); (M.T.R.-Á.)
| |
Collapse
|
22
|
Ferng D, Sun W, Shieh BH. Differential activation of rhodopsin triggers distinct endocytic trafficking and recycling in vivo via differential phosphorylation. PLoS One 2024; 19:e0303882. [PMID: 38848405 PMCID: PMC11161057 DOI: 10.1371/journal.pone.0303882] [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: 01/03/2024] [Accepted: 05/02/2024] [Indexed: 06/09/2024] Open
Abstract
Activated GPCRs are phosphorylated and internalized mostly via clathrin-mediated endocytosis (CME), which are then sorted for recycling or degradation. We investigated how differential activation of the same GPCR affects its endocytic trafficking in vivo using rhodopsin as a model in pupal photoreceptors of flies expressing mCherry-tagged rhodopsin 1 (Rh1-mC) or GFP-tagged arrestin 1 (Arr1-GFP). Upon blue light stimulation, activated Rh1 recruited Arr1-GFP to the rhabdomere, which became co-internalized and accumulated in cytoplasmic vesicles of photoreceptors. This internalization was eliminated in shits1 mutants affecting dynamin. Moreover, it was blocked by either rdgA or rdgB mutations affecting the PIP2 biosynthesis. Together, the blue light-initiated internalization of Rh1 and Arr1 belongs to CME. Green light stimulation also triggered the internalization and accumulation of activated Rh1-mC in the cytoplasm but with faster kinetics. Importantly, Arr1-GFP was also recruited to the rhabdomere but not co-internalized with Rh1-mC. This endocytosis was not affected in shits1 nor rdgA mutants, indicating it is not CME. We explored the fate of internalized Rh1-mC following CME and observed it remained in cytoplasmic vesicles following 30 min of dark adaptation. In contrast, in the non-CME Rh1-mC appeared readily recycled back to the rhabdomere within five min of dark treatment. This faster recycling may be regulated by rhodopsin phosphatase, RdgC. Together, we demonstrate two distinct endocytic and recycling mechanisms of Rh1 via two light stimulations. It appears that each stimulation triggers a distinct conformation leading to different phosphorylation patterns of Rh1 capable of recruiting Arr1 to rhabdomeres. However, a more stable interaction leads to the co-internalization of Arr1 that orchestrates CME. A stronger Arr1 association appears to impede the recycling of the phosphorylated Rh1 by preventing the recruitment of RdgC. We conclude that conformations of activated rhodopsin determine the downstream outputs upon phosphorylation that confers differential protein-protein interactions.
Collapse
Affiliation(s)
- Darwin Ferng
- Department of Pharmacology, Center for Molecular Neuroscience and Vision Research Center, Vanderbilt University, Nashville, TN, United States of America
| | - Wesley Sun
- Department of Pharmacology, Center for Molecular Neuroscience and Vision Research Center, Vanderbilt University, Nashville, TN, United States of America
| | - Bih-Hwa Shieh
- Department of Pharmacology, Center for Molecular Neuroscience and Vision Research Center, Vanderbilt University, Nashville, TN, United States of America
| |
Collapse
|
23
|
Simmons SR, Herring SE, Tchalla EYI, Lenhard AP, Bhalla M, Bou Ghanem EN. Activating A1 adenosine receptor signaling boosts early pulmonary neutrophil recruitment in aged mice in response to Streptococcus pneumoniae infection. Immun Ageing 2024; 21:34. [PMID: 38840213 PMCID: PMC11151497 DOI: 10.1186/s12979-024-00442-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Streptococcus pneumoniae (pneumococcus) is a leading cause of pneumonia in older adults. Successful control of pneumococci requires robust pulmonary neutrophil influx early in infection. However, aging is associated with aberrant neutrophil recruitment and the mechanisms behind that are not understood. Here we explored how neutrophil recruitment following pneumococcal infection changes with age and the host pathways regulating this. RESULTS Following pneumococcal infection there was a significant delay in early neutrophil recruitment to the lungs of aged mice. Neutrophils from aged mice showed defects in trans-endothelial migration in vitro compared to young controls. To understand the pathways involved, we examined immune modulatory extracellular adenosine (EAD) signaling, that is activated upon cellular damage. Signaling through the lower affinity A2A and A2B adenosine receptors had no effect on neutrophil recruitment to infected lungs. In contrast, inhibition of the high affinity A1 receptor in young mice blunted neutrophil recruitment to the lungs following infection. A1 receptor inhibition decreased expression of CXCR2 on circulating neutrophils, which is required for trans-endothelial migration. Indeed, A1 receptor signaling on neutrophils was required for their ability to migrate across endothelial cells in response to infection. Aging was not associated with defects in EAD production or receptor expression on neutrophils. However, agonism of A1 receptor in aged mice rescued the early defect in neutrophil migration to the lungs and improved control of bacterial burden. CONCLUSIONS This study suggests age-driven defects in EAD damage signaling can be targeted to rescue the delay in pulmonary neutrophil migration in response to bacterial pneumonia.
Collapse
Affiliation(s)
- Shaunna R Simmons
- Department of Microbiology and Immunology, School of Medicine, University at Buffalo, Buffalo, NY, USA
| | - Sydney E Herring
- Department of Microbiology and Immunology, School of Medicine, University at Buffalo, Buffalo, NY, USA
| | - Essi Y I Tchalla
- Department of Microbiology and Immunology, School of Medicine, University at Buffalo, Buffalo, NY, USA
| | - Alexsandra P Lenhard
- Department of Microbiology and Immunology, School of Medicine, University at Buffalo, Buffalo, NY, USA
| | - Manmeet Bhalla
- Department of Microbiology and Immunology, School of Medicine, University at Buffalo, Buffalo, NY, USA
| | - Elsa N Bou Ghanem
- Department of Microbiology and Immunology, School of Medicine, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
24
|
Cheng N, Pimentel JM, Trejo J. Ubiquitin-driven G protein-coupled receptor inflammatory signaling at the endosome. Am J Physiol Cell Physiol 2024; 326:C1605-C1610. [PMID: 38646783 PMCID: PMC11371321 DOI: 10.1152/ajpcell.00161.2024] [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: 03/10/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/23/2024]
Abstract
G protein-coupled receptors (GPCRs) are ubiquitously expressed cell surface receptors that mediate numerous physiological responses and are highly druggable. Upon activation, GPCRs rapidly couple to heterotrimeric G proteins and are then phosphorylated and internalized from the cell surface. Recent studies indicate that GPCRs not only localize at the plasma membrane but also exist in intracellular compartments where they are competent to signal. Intracellular signaling by GPCRs is best described to occur at endosomes. Several studies have elegantly documented endosomal GPCR-G protein and GPCR-β-arrestin signaling. Besides phosphorylation, GPCRs are also posttranslationally modified with ubiquitin. GPCR ubiquitination has been studied mainly in the context of receptor endosomal-lysosomal trafficking. However, new studies indicate that ubiquitination of endogenous GPCRs expressed in endothelial cells initiates the assembly of an intracellular p38 mitogen-activated kinase signaling complex that promotes inflammatory responses from endosomes. In this mini-review, we discuss emerging discoveries that provide critical insights into the function of ubiquitination in regulating GPCR inflammatory signaling at endosomes.
Collapse
Affiliation(s)
- Norton Cheng
- Department of Pharmacology, School of Medicine, University of California, San Diego, California, United States
- Biomedical Sciences Graduate Program, University of California, San Diego, California, United States
| | - Julio M Pimentel
- Department of Pharmacology, School of Medicine, University of California, San Diego, California, United States
| | - JoAnn Trejo
- Department of Pharmacology, School of Medicine, University of California, San Diego, California, United States
| |
Collapse
|
25
|
Forsman H, Dahlgren C, Mårtensson J, Björkman L, Sundqvist M. Function and regulation of GPR84 in human neutrophils. Br J Pharmacol 2024; 181:1536-1549. [PMID: 36869866 DOI: 10.1111/bph.16066] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023] Open
Abstract
Human neutrophils are components of the innate immune system and are the most abundant white blood cells in the circulation. They are professional phagocytes and express several G protein-coupled receptors (GPCRs), which are essential for proper neutrophil functions. So far, the two formyl peptide receptors, FPR1 and FPR2, have been the most extensively studied group of neutrophil GPCRs, but recently, a new group, the free fatty acid (FFA) receptors, has attracted growing attention. Neutrophils express two FFA receptors, GPR84 and FFA2, which sense medium- and short-chain fatty acids respectively, and display similar activation profiles. The exact pathophysiological role of GPR84 is not yet fully understood, but it is generally regarded as a pro-inflammatory receptor that mediates neutrophil activation. In this review, we summarize current knowledge of how GPR84 affects human neutrophil functions and discuss the regulatory mechanisms that control these responses, focusing on the similarities and differences in comparison to the two FPRs and FFA2. LINKED ARTICLES: This article is part of a themed issue GPR84 Pharmacology. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.10/issuetoc.
Collapse
Affiliation(s)
- Huamei Forsman
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Claes Dahlgren
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jonas Mårtensson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lena Björkman
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Martina Sundqvist
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
26
|
Durydivka O, Palivec P, Gazdarica M, Mackie K, Blahos J, Kuchar M. Hexahydrocannabinol (HHC) and Δ 9-tetrahydrocannabinol (Δ 9-THC) driven activation of cannabinoid receptor 1 results in biased intracellular signaling. Sci Rep 2024; 14:9181. [PMID: 38649680 PMCID: PMC11035541 DOI: 10.1038/s41598-024-58845-7] [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: 11/21/2023] [Accepted: 04/03/2024] [Indexed: 04/25/2024] Open
Abstract
The Cannabis sativa plant has been used for centuries as a recreational drug and more recently in the treatment of patients with neurological or psychiatric disorders. In many instances, treatment goals include relief from posttraumatic disorders, anxiety, or to support treatment of chronic pain. Ligands acting on cannabinoid receptor 1 (CB1R) are also potential targets for the treatment of other health conditions. Using an evidence-based approach, pharmacological investigation of CB1R agonists is timely, with the aim to provide chronically ill patients relief using well-defined and characterized compounds from cannabis. Hexahydrocannabinol (HHC), currently available over the counter in many countries to adults and even children, is of great interests to policy makers, legal administrators, and healthcare regulators, as well as pharmacologists. Herein, we studied the pharmacodynamics of HHC epimers, which activate CB1R. We compared their key CB1R-mediated signaling pathway activities and compared them to the pathways activated by Δ9-tetrahydrocannabinol (Δ9-THC). We provide evidence that activation of CB1R by HHC ligands is only broadly comparable to those mediated by Δ9-THC, and that both HHC epimers have unique properties. Together with the greater chemical stability of HHC compared to Δ9-THC, these molecules have a potential to become a part of modern medicine.
Collapse
Affiliation(s)
- Oleh Durydivka
- Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague 4, Czech Republic.
- Forensic Laboratory of Biologically Active Substances, Department of Chemistry of Natural Compounds, University of Chemistry and Technology Prague, Technicka 3, Prague, Czech Republic.
| | - Petr Palivec
- Forensic Laboratory of Biologically Active Substances, Department of Chemistry of Natural Compounds, University of Chemistry and Technology Prague, Technicka 3, Prague, Czech Republic
| | - Matej Gazdarica
- Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Ken Mackie
- Department of Psychological and Brain Sciences, Gill Center for Molecular Bioscience, Indiana University, 1101 E. 10th St., Bloomington, IN, 47405, USA
| | - Jaroslav Blahos
- Institute of Molecular Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20, Prague 4, Czech Republic
| | - Martin Kuchar
- Forensic Laboratory of Biologically Active Substances, Department of Chemistry of Natural Compounds, University of Chemistry and Technology Prague, Technicka 3, Prague, Czech Republic.
- Psychedelic Research Center, National Institute of Mental Health, Topolová 748, Klecany, Czech Republic.
| |
Collapse
|
27
|
Simmons SR, Herring SE, Tchalla EYI, Lenhard AP, Bhalla M, Bou Ghanem EN. Activating A1 adenosine receptor signaling boosts early pulmonary neutrophil recruitment in aged mice in response to Streptococcus pneumoniae infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.08.574741. [PMID: 38260350 PMCID: PMC10802397 DOI: 10.1101/2024.01.08.574741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background Streptococcus pneumoniae (pneumococcus) is a leading cause of pneumonia in older adults. Successful control of pneumococci requires robust pulmonary neutrophil influx early in infection. However, aging is associated with aberrant neutrophil recruitment and the mechanisms behind that are not understood. Here we explored how neutrophil recruitment following pneumococcal infection changes with age and the host pathways regulating this. Results Following pneumococcal infection there was a significant delay in early neutrophil recruitment to the lungs of aged mice. Neutrophils from aged mice showed defects in trans-endothelial migration in vitro compared to young controls. To understand the pathways involved, we examined immune modulatory extracellular adenosine (EAD) signaling, that is activated upon cellular damage. Signaling through the lower affinity A2A and A2B adenosine receptors had no effect on neutrophil recruitment to infected lungs. In contrast, inhibition of the high affinity A1 receptor in young mice blunted neutrophil recruitment to the lungs following infection. A1 receptor inhibition decreased expression of CXCR2 on circulating neutrophils, which is required for transendothelial migration. Indeed, A1 receptor signaling on neutrophils was required for their ability to migrate across endothelial cells in response to infection. Aging was not associated with defects in EAD production or receptor expression on neutrophils. However, agonism of A1 receptor in aged mice rescued the early defect in neutrophil migration to the lungs and improved control of bacterial burden. Conclusions This study suggests age-driven defects in EAD damage signaling can be targeted to rescue the delay in pulmonary neutrophil migration in response to bacterial pneumonia.
Collapse
|
28
|
Ke P, Zhu DN, Liu MZ, Yan H, Zhao QJ, Du J, Wei W, Chen XW, Liu C. Macrophage β-arrestin-1 deteriorates DSS-induced colitis through interaction with NF-κB signaling. Int Immunopharmacol 2024; 130:111676. [PMID: 38367465 DOI: 10.1016/j.intimp.2024.111676] [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: 10/22/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/19/2024]
Abstract
β-arrestin-1 has been demonstrated to participate in the regulation of inflammatory reactions in several diseases. Thus, this study aimed to investigate the role of macrophage β-arrestin-1 in the pathogenesis and progression of ulcerative colitis (UC). A myeloid β-arrestin-1 conditional knockout mouse model was generated to explore the role of macrophage β-arrestin-1. DSS was employed for the establishment of an ulcerative colitis mouse model, using TNF-α as an inflammatory stressor in vitro. The expression level of β-arrestin-1 was detected via western blot and immunofluorescence assays, whilst disease severity was evaluated by clinical score and H&E staining in the DSS-induced colitis model. In the in vitro experiments, the levels of inflammatory cytokines were examined using real-time PCR. NF-κB activation was detected through the double luciferase reporter system, western blot, and electrophoretic mobility shift assay (EMSA). BAY11-7082 was used to inhibit NF-κB activation. Our results exposed that the level of β-arrestin-1 was increased in monocytes/macrophages derived from DSS-induced colitis mice or under the TNF-α challenge. Moreover, conditionally knocking out the expression of myeloid β-arrestin-1 alleviated disease severity, while knocking out the expression of β-arrestin-1 decreased the levels of inflammatory cytokines. Additionally, NF-κB was identified as a central regulatory element of β-arrestin-1 promoter, and using BAY11-7082 to inhibit NF-κB activation lowered the level of β-arrestin-1 under TNF-α challenge. β-arrestin-1 led to the activation of the NF-κB signaling pathway by enhancing binding to IκBα and IKK under the TNF-α challenge. Taken together, our findings demonstrated macrophage β-arrestin-1 contributes to the deterioration of DSS-induced colitis through the interaction with NF-κB signaling, thus highlighting a novel target for the treatment of UC.
Collapse
Affiliation(s)
- Ping Ke
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 20043 China; Air Force Hangzhou Special Service Recuperation Center, Sanatorium Area 4, Nanjing 211131 China
| | - Dan-Ni Zhu
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 20043 China
| | - Meng-Zhen Liu
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 20043 China
| | - Hui Yan
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 20043 China
| | - Qing-Jie Zhao
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 20043 China
| | - Jing Du
- Department of Basic Medicine, School of Pharmacology, Jining Medical University, Jining 272067 China
| | - Wei Wei
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Xiong-Wen Chen
- Department of Biopharmaceuticals & Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Heping District, Tianjin, China
| | - Chong Liu
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 20043 China.
| |
Collapse
|
29
|
Lattanzi R, Casella I, Fullone MR, Maftei D, Vincenzi M, Miele R. MRAP2 Inhibits β-Arrestin-2 Recruitment to the Prokineticin Receptor 2. Curr Issues Mol Biol 2024; 46:1607-1620. [PMID: 38392222 PMCID: PMC10887741 DOI: 10.3390/cimb46020104] [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: 12/22/2023] [Revised: 02/05/2024] [Accepted: 02/14/2024] [Indexed: 02/24/2024] Open
Abstract
Melanocortin receptor accessory protein 2 (MRAP2) is a membrane protein that binds multiple G protein-coupled receptors (GPCRs) involved in the control of energy homeostasis, including prokineticin receptors. These GPCRs are expressed both centrally and peripherally, and their endogenous ligands are prokineticin 1 (PK1) and prokineticin 2 (PK2). PKRs couple all G-protein subtypes, such as Gαq/11, Gαs, and Gαi, and recruit β-arrestins upon PK2 stimulation, although the interaction between PKR2 and β-arrestins does not trigger receptor internalisation. MRAP2 inhibits the anorexigenic effect of PK2 by binding PKR1 and PKR2. The aim of this work was to elucidate the role of MRAP2 in modulating PKR2-induced β-arrestin-2 recruitment and β-arrestin-mediated signalling. This study could allow the identification of new specific targets for potential new drugs useful for the treatment of the various pathologies correlated with prokineticin, in particular, obesity.
Collapse
Affiliation(s)
- Roberta Lattanzi
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (R.L.); (D.M.); (M.V.)
| | - Ida Casella
- National Centre for Drug Research and Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Maria Rosaria Fullone
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy;
| | - Daniela Maftei
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (R.L.); (D.M.); (M.V.)
| | - Martina Vincenzi
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (R.L.); (D.M.); (M.V.)
| | - Rossella Miele
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy;
| |
Collapse
|
30
|
Nibley PC, Shenoy SK. β-adrenergic receptor signaling mediated by β-arrestins and its potential role in heart failure. CURRENT OPINION IN PHYSIOLOGY 2024; 37:100723. [PMID: 38094036 PMCID: PMC10715791 DOI: 10.1016/j.cophys.2023.100723] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
Abstract
The lethality of heart failure (HF), particularly in the context of post-acute sequelae SARS-CoV-2 infection (PASC)-related myocarditis, necessitates the discovery of the cellular pathways implicated in cardiovascular disease (CVD). We summarize the signaling mechanisms of the catecholamine-binding β-adrenergic receptors (β-ARs), with an emphasis on the role of β-arrestins. β-ARs, a subset of G protein-coupled receptors (GPCRs), canonically propagate signals through heterotrimeric G proteins. However, since their discovery in the late 1980s, β-arrestins have been shown to, both (i) quench G protein signaling and (ii) initiate their own independent signaling cascades, which is influenced by post-translational modifications. β-arrestin-biased agonism by the beta-blocker carvedilol and its allosteric modulation can serve a cardioprotective role. The increasingly labyrinthine nature of GPCR signaling suggests that ligand-dependent β-AR signaling, either stimulated by an agonist or blocked by an antagonist, is selectively enhanced or suppressed by allosteric modulations, which are orchestrated by novel drugs or endogenous post-translational modifications.
Collapse
Affiliation(s)
- Preston C. Nibley
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Sudha K. Shenoy
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
31
|
Kee TR, Khan SA, Neidhart MB, Masters BM, Zhao VK, Kim YK, McGill Percy KC, Woo JAA. The multifaceted functions of β-arrestins and their therapeutic potential in neurodegenerative diseases. Exp Mol Med 2024; 56:129-141. [PMID: 38212557 PMCID: PMC10834518 DOI: 10.1038/s12276-023-01144-4] [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: 04/20/2023] [Revised: 10/18/2023] [Accepted: 10/20/2023] [Indexed: 01/13/2024] Open
Abstract
Arrestins are multifunctional proteins that regulate G-protein-coupled receptor (GPCR) desensitization, signaling, and internalization. The arrestin family consists of four subtypes: visual arrestin1, β-arrestin1, β-arrestin2, and visual arrestin-4. Recent studies have revealed the multifunctional roles of β-arrestins beyond GPCR signaling, including scaffolding and adapter functions, and physically interacting with non-GPCR receptors. Increasing evidence suggests that β-arrestins are involved in the pathogenesis of a variety of neurodegenerative diseases, including Alzheimer's disease (AD), frontotemporal dementia (FTD), and Parkinson's disease (PD). β-arrestins physically interact with γ-secretase, leading to increased production and accumulation of amyloid-beta in AD. Furthermore, β-arrestin oligomers inhibit the autophagy cargo receptor p62/SQSTM1, resulting in tau accumulation and aggregation in FTD. In PD, β-arrestins are upregulated in postmortem brain tissue and an MPTP model, and the β2AR regulates SNCA gene expression. In this review, we aim to provide an overview of β-arrestin1 and β-arrestin2, and describe their physiological functions and roles in neurodegenerative diseases. The multifaceted roles of β-arrestins and their involvement in neurodegenerative diseases suggest that they may serve as promising therapeutic targets.
Collapse
Affiliation(s)
- Teresa R Kee
- Department of Pathology, CWRU School of Medicine, Cleveland, OH, 44106, USA
- Department of Molecular Medicine, USF Health College of Medicine, Tampa, FL, 33613, USA
| | - Sophia A Khan
- Department of Pathology, CWRU School of Medicine, Cleveland, OH, 44106, USA
| | - Maya B Neidhart
- Department of Pathology, CWRU School of Medicine, Cleveland, OH, 44106, USA
| | - Brianna M Masters
- Department of Pathology, CWRU School of Medicine, Cleveland, OH, 44106, USA
| | - Victoria K Zhao
- Department of Pathology, CWRU School of Medicine, Cleveland, OH, 44106, USA
| | - Yenna K Kim
- Department of Pathology, CWRU School of Medicine, Cleveland, OH, 44106, USA
| | | | - Jung-A A Woo
- Department of Pathology, CWRU School of Medicine, Cleveland, OH, 44106, USA.
| |
Collapse
|
32
|
Lee KT, Pranoto IKA, Kim SY, Choi HJ, To NB, Chae H, Lee JY, Kim JE, Kwon YV, Nam JW. Comparative interactome analysis of α-arrestin families in human and Drosophila. eLife 2024; 12:RP88328. [PMID: 38270169 PMCID: PMC10945707 DOI: 10.7554/elife.88328] [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: 01/26/2024] Open
Abstract
The α-arrestins form a large family of evolutionally conserved modulators that control diverse signaling pathways, including both G-protein-coupled receptor (GPCR)-mediated and non-GPCR-mediated pathways, across eukaryotes. However, unlike β-arrestins, only a few α-arrestin targets and functions have been characterized. Here, using affinity purification and mass spectrometry, we constructed interactomes for 6 human and 12 Drosophila α-arrestins. The resulting high-confidence interactomes comprised 307 and 467 prey proteins in human and Drosophila, respectively. A comparative analysis of these interactomes predicted not only conserved binding partners, such as motor proteins, proteases, ubiquitin ligases, RNA splicing factors, and GTPase-activating proteins, but also those specific to mammals, such as histone modifiers and the subunits of V-type ATPase. Given the manifestation of the interaction between the human α-arrestin, TXNIP, and the histone-modifying enzymes, including HDAC2, we undertook a global analysis of transcription signals and chromatin structures that were affected by TXNIP knockdown. We found that TXNIP activated targets by blocking HDAC2 recruitment to targets, a result that was validated by chromatin immunoprecipitation assays. Additionally, the interactome for an uncharacterized human α-arrestin ARRDC5 uncovered multiple components in the V-type ATPase, which plays a key role in bone resorption by osteoclasts. Our study presents conserved and species-specific protein-protein interaction maps for α-arrestins, which provide a valuable resource for interrogating their cellular functions for both basic and clinical research.
Collapse
Affiliation(s)
- Kyung-Tae Lee
- Department of Life Science, College of Natural Sciences, Hanyang UniversitySeoulRepublic of Korea
- Hanyang Institute of Advanced BioConvergence, Hanyang UniversitySeoulRepublic of Korea
| | - Inez KA Pranoto
- Department of Biochemistry, University of WashingtonSeattleUnited States
| | - Soon-Young Kim
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National UniversityDaeguRepublic of Korea
| | - Hee-Joo Choi
- Bio-BigData Center, Hanyang Institute for Bioscience and Biotechnology, Hanyang UniversitySeoulRepublic of Korea
- Department of Pathology, College of Medicine, Hanyang UniversitySeoulRepublic of Korea
- Hanyang Biomedical Research Institute, Hanyang UniversitySeoulRepublic of Korea
| | - Ngoc Bao To
- Department of Life Science, College of Natural Sciences, Hanyang UniversitySeoulRepublic of Korea
| | - Hansong Chae
- Department of Life Science, College of Natural Sciences, Hanyang UniversitySeoulRepublic of Korea
| | - Jeong-Yeon Lee
- Bio-BigData Center, Hanyang Institute for Bioscience and Biotechnology, Hanyang UniversitySeoulRepublic of Korea
- Department of Pathology, College of Medicine, Hanyang UniversitySeoulRepublic of Korea
| | - Jung-Eun Kim
- Department of Molecular Medicine, Cell and Matrix Research Institute, School of Medicine, Kyungpook National UniversityDaeguRepublic of Korea
| | - Young V Kwon
- Department of Biochemistry, University of WashingtonSeattleUnited States
| | - Jin-Wu Nam
- Department of Life Science, College of Natural Sciences, Hanyang UniversitySeoulRepublic of Korea
- Hanyang Institute of Advanced BioConvergence, Hanyang UniversitySeoulRepublic of Korea
- Bio-BigData Center, Hanyang Institute for Bioscience and Biotechnology, Hanyang UniversitySeoulRepublic of Korea
| |
Collapse
|
33
|
Lohse MJ, Bock A, Zaccolo M. G Protein-Coupled Receptor Signaling: New Insights Define Cellular Nanodomains. Annu Rev Pharmacol Toxicol 2024; 64:387-415. [PMID: 37683278 DOI: 10.1146/annurev-pharmtox-040623-115054] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
G protein-coupled receptors are the largest and pharmacologically most important receptor family and are involved in the regulation of most cell functions. Most of them reside exclusively at the cell surface, from where they signal via heterotrimeric G proteins to control the production of second messengers such as cAMP and IP3 as well as the activity of several ion channels. However, they may also internalize upon agonist stimulation or constitutively reside in various intracellular locations. Recent evidence indicates that their function differs depending on their precise cellular localization. This is because the signals they produce, notably cAMP and Ca2+, are mostly bound to cell proteins that significantly reduce their mobility, allowing the generation of steep concentration gradients. As a result, signals generated by the receptors remain confined to nanometer-sized domains. We propose that such nanometer-sized domains represent the basic signaling units in a cell and a new type of target for drug development.
Collapse
Affiliation(s)
- Martin J Lohse
- ISAR Bioscience Institute, Planegg/Munich, Germany;
- Rudolf Boehm Institute of Pharmacology and Toxicology, Leipzig University, Leipzig, Germany
| | - Andreas Bock
- Rudolf Boehm Institute of Pharmacology and Toxicology, Leipzig University, Leipzig, Germany
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics and National Institute for Health and Care Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom;
| |
Collapse
|
34
|
Faivre N, Verollet C, Dumas F. The chemokine receptor CCR5: multi-faceted hook for HIV-1. Retrovirology 2024; 21:2. [PMID: 38263120 PMCID: PMC10807162 DOI: 10.1186/s12977-024-00634-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 01/13/2024] [Indexed: 01/25/2024] Open
Abstract
Chemokines are cytokines whose primary role is cellular activation and stimulation of leukocyte migration. They perform their various functions by interacting with G protein-coupled cell surface receptors (GPCRs) and are involved in the regulation of many biological processes such as apoptosis, proliferation, angiogenesis, hematopoiesis or organogenesis. They contribute to the maintenance of the homeostasis of lymphocytes and coordinate the function of the immune system. However, chemokines and their receptors are sometimes hijacked by some pathogens to infect the host organism. For a given chemokine receptor, there is a wide structural, organizational and conformational diversity. In this review, we describe the evidence for structural variety reported for the chemokine receptor CCR5, how this variability can be exploited by HIV-1 to infect its target cells and what therapeutic solutions are currently being developed to overcome this problem.
Collapse
Affiliation(s)
- Natacha Faivre
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
- International Research Laboratory (IRP) CNRS "IM-TB/HIV", Toulouse, France
- International Research Laboratory (IRP) CNRS "IM-TB/HIV", Buenos Aires, Argentina
| | - Christel Verollet
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France
- International Research Laboratory (IRP) CNRS "IM-TB/HIV", Toulouse, France
- International Research Laboratory (IRP) CNRS "IM-TB/HIV", Buenos Aires, Argentina
| | - Fabrice Dumas
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UPS), Toulouse, France.
| |
Collapse
|
35
|
Møller TC, Moo EV, Inoue A, Pedersen MF, Bräuner-Osborne H. Characterization of the real-time internalization of nine GPCRs reveals distinct dependence on arrestins and G proteins. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119584. [PMID: 37714305 DOI: 10.1016/j.bbamcr.2023.119584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/24/2023] [Accepted: 09/06/2023] [Indexed: 09/17/2023]
Abstract
G protein-coupled receptors (GPCRs) are seven transmembrane receptors that respond to external stimuli and undergo conformational changes to activate G proteins and modulate cellular processes leading to biological outcomes. To prevent overstimulation and prolonged exposure to stimuli, GPCRs are regulated by internalization. While the canonical GPCR internalization mechanism in mammalian cells is arrestin-dependent, clathrin-mediated endocytosis, more diverse GPCR internalization mechanisms have been described over the years. However, there is a lack of consistent methods used in the literature making it complicated to determine a receptor's internalization pathway. Here, we utilized a highly efficient time-resolved Förster resonance energy transfer (TR-FRET) internalization assay to determine the internalization profile of nine distinct GPCRs representing the GPCR classes A, B and C and with different G protein coupling profiles. This technique, coupled with clustered regularly interspaced palindromic repeats (CRISPR) engineered knockout cells allows us to effectively study the involvement of heterotrimeric G proteins and non-visual arrestins. We found that all the nine receptors internalized upon agonist stimulation in a concentration-dependent manner and six receptors showed basal internalization. Yet, there is no correlation between the receptor class and primary G protein coupling to the arrestin and G protein dependence for GPCR internalization. Overall, this study presents a platform for studying internalization that is applicable to most GPCRs and may even be extended to other membrane proteins. This method can be easily applicable to other endocytic machinery of interest and ultimately will lend itself towards the construction of comprehensive receptor internalization profiles.
Collapse
Affiliation(s)
- Thor C Møller
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Ee Von Moo
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark.
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Mie F Pedersen
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Hans Bräuner-Osborne
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark
| |
Collapse
|
36
|
Mastos C, Xu X, Keen AC, Halls ML. Signalling of Adrenoceptors: Canonical Pathways and New Paradigms. Handb Exp Pharmacol 2024; 285:147-184. [PMID: 38227198 DOI: 10.1007/164_2023_704] [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: 01/17/2024]
Abstract
The concept of G protein-coupled receptors initially arose from studies of the β-adrenoceptor, adenylyl cyclase, and cAMP signalling pathway. Since then both canonical G protein-coupled receptor signalling pathways and emerging paradigms in receptor signalling have been defined by experiments focused on adrenoceptors. Here, we discuss the evidence for G protein coupling specificity of the nine adrenoceptor subtypes. We summarise the ability of each of the adrenoceptors to activate proximal signalling mediators including cAMP, calcium, mitogen-activated protein kinases, and protein kinase C pathways. Finally, we highlight the importance of precise spatial and temporal control of adrenoceptor signalling that is controlled by the localisation of receptors at intracellular membranes and in larger protein complexes.
Collapse
Affiliation(s)
- Chantel Mastos
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Xiaomeng Xu
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Alastair C Keen
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Michelle L Halls
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.
| |
Collapse
|
37
|
Akkenepally SV, Yombo DJK, Yerubandi S, Reddy GB, Deshpande DA, McCormack FX, Madala SK. Interleukin 31 receptor α promotes smooth muscle cell contraction and airway hyperresponsiveness in asthma. Nat Commun 2023; 14:8207. [PMID: 38081868 PMCID: PMC10713652 DOI: 10.1038/s41467-023-44040-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Asthma is a chronic inflammatory airway disease characterized by airway hyperresponsiveness (AHR), inflammation, and goblet cell hyperplasia. Multiple cytokines, including IFNγ, IL-4, and IL-13 are associated with asthma; however, the mechanisms underlying the effects of these cytokines remain unclear. Here, we report a significant increase in the expression of IL-31RA, but not its cognate ligand IL-31, in mouse models of allergic asthma. In support of this, IFNγ, IL-4, and IL-13 upregulated IL-31RA but not IL-31 in both human and mice primary airway smooth muscle cells (ASMC) isolated from the airways of murine and human lungs. Importantly, the loss of IL-31RA attenuated AHR but had no effect on inflammation and goblet cell hyperplasia in mice challenged with allergens or treated with IL-13 or IFNγ. We show that IL-31RA functions as a positive regulator of muscarinic acetylcholine receptor 3 expression, augmenting calcium levels and myosin light chain phosphorylation in human and murine ASMC. These findings identify a role for IL-31RA in AHR that is distinct from airway inflammation and goblet cell hyperplasia in asthma.
Collapse
Affiliation(s)
- Santhoshi V Akkenepally
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
- Division of Biochemistry, National Institute of Nutrition, Hyderabad, Telangana, India
| | - Dan J K Yombo
- Division of Pulmonary Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sanjana Yerubandi
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
- Division of Pulmonary Medicine, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | - Deepak A Deshpande
- Division of Pulmonary, Allergy, and Critical Care Medicine, Center for Translational Medicine, Jane and Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Francis X McCormack
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Satish K Madala
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
38
|
He Y, Su Q, Zhao L, Zhang L, Yu L, Shi J. Historical perspectives and recent advances in small molecule ligands of selective/biased/multi-targeted μ/δ/κ opioid receptor (2019-2022). Bioorg Chem 2023; 141:106869. [PMID: 37797454 DOI: 10.1016/j.bioorg.2023.106869] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/11/2023] [Accepted: 09/15/2023] [Indexed: 10/07/2023]
Abstract
The opioids have been used for more than a thousand years and are not only the most widely prescribed drugs for moderate to severe pain and acute pain, but also the preferred drugs. However, their non-analgesic effects, especially respiratory depression and potential addiction, are important factors that plague the safety of clinical use and are an urgent problem for pharmacological researchers to address. Current research on analgesic drugs has evolved into different directions: de-opioidization; application of pharmacogenomics to individualize the use of opioids; development of new opioids with less adverse effects. The development of new opioid drugs remains a hot research topic, and with the in-depth study of opioid receptors and intracellular signal transduction mechanisms, new research ideas have been provided for the development of new opioid analgesics with less side effects and stronger analgesic effects. The development of novel opioid drugs in turn includes selective opioid receptor ligands, biased opioid receptor ligands, and multi-target opioid receptor ligands and positive allosteric modulators (PAMs) or antagonists and the single compound as multi-targeted agnoists/antagonists for different receptors. PAMs strategies are also getting newer and are the current research hotspots, including the BMS series of compounds and others, which are extensive and beyond the scope of this review. This review mainly focuses on the selective/biased/multi-targeted MOR/DOR/KOR (mu opioid receptor/delta opioid receptor/kappa opioid receptor) small molecule ligands and involves some cryo-electron microscopy (cryoEM) and structure-based approaches as well as the single compound as multi-targeted agnoists/antagonists for different receptors from 2019 to 2022, including discovery history, activities in vitro and vivo, and clinical studies, in an attempt to provide ideas for the development of novel opioid analgesics with fewer side effects.
Collapse
Affiliation(s)
- Ye He
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Qian Su
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Liyun Zhao
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Lijuan Zhang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| | - Lu Yu
- Department of Respiratory Medicine, Sichuan Academy of Medical Sciences and Sichuan provincial People's Hospital, Chengdu, 610072, China.
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China; State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China.
| |
Collapse
|
39
|
Mandeville JB, Weigand-Whittier J, Wey HY, Chen YCI. Amphetamine pretreatment blunts dopamine-induced D2/D3-receptor occupancy by an arrestin-mediated mechanism: A PET study in internalization compromised mice. Neuroimage 2023; 283:120416. [PMID: 37866759 PMCID: PMC10841768 DOI: 10.1016/j.neuroimage.2023.120416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/29/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023] Open
Abstract
While all reversible receptor-targeting radioligands for positron emission tomography (PET) can be displaced by competition with an antagonist at the receptor, many radiotracers show limited occupancies using agonists even at high doses. [11C]Raclopride, a D2/D3 receptor radiotracer with rapid kinetics, can identify the direction of changes in the neurotransmitter dopamine, but quantitative interpretation of the relationship between dopamine levels and radiotracer binding has proven elusive. Agonist-induced receptor desensitization and internalization, a homeostatic mechanism to downregulate neurotransmitter-mediated function, can shift radioligand-receptor binding affinity and confound PET interpretations of receptor occupancy. In this study, we compared occupancies induced by amphetamine (AMP) in drug-naive wild-type (WT) and internalization-compromised β-arrestin-2 knockout (KO) mice using a within-scan drug infusion to modulate the kinetics of [11C]raclopride. We additionally performed studies at 3 h following AMP pretreatment, with the hypothesis that receptor internalization should markedly attenuate occupancy on the second challenge, because dopamine cannot access internalized receptors. Without prior AMP treatment, WT mice exhibited somewhat larger binding potential than KO mice but similar AMP-induced occupancy. At 3 h after AMP treatment, WT mice exhibited binding potentials that were 15 % lower than KO mice. At this time point, occupancy was preserved in KO mice but suppressed by 60 % in WT animals, consistent with a model in which most receptors contributing to binding potential in WT animals were not functional. These results demonstrate that arrestin-mediated receptor desensitization and internalization produce large effects in PET [11C]raclopride occupancy studies using agonist challenges.
Collapse
Affiliation(s)
- Joseph B Mandeville
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Jonah Weigand-Whittier
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA; Department of Bioengineering, University of California, Berkeley, CA, USA
| | - Hsiao-Ying Wey
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Yin-Ching I Chen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| |
Collapse
|
40
|
Shao BZ, Liu MZ, Zhu DN, Yan H, Ke P, Wei W, Han T, Liu C. Depletion of β-arrestin-1 in macrophages enhances atherosclerosis in ApoE -/- mice. Int Immunopharmacol 2023; 125:111085. [PMID: 37866313 DOI: 10.1016/j.intimp.2023.111085] [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: 07/12/2023] [Revised: 10/12/2023] [Accepted: 10/15/2023] [Indexed: 10/24/2023]
Abstract
Autophagy in atherosclerotic plaque macrophage contributes to the alleviation of atherosclerosis through the promotion of lipid metabolism. β-arrestins are multifunctional proteins participating various kinds of cellular signaling pathways. Here we aimed to determine the role of β-arrestin-1, an important member of β-arrestin family, in atherosclerosis, and whether autophagy was involved in this process. ApoE-/-β-arrestin-1fl/flLysM-Cre mice were created through bone marrow transplantation for the atherosclerosis model with conditional myeloid knocking out β-arrestin-1. Bone marrow-derived macrophages (BMDMs) were used for the in vitro studies. Oil red O staining was used to detect the lesional area. F4/80, Masson trichrome and picro-Sirius red staining were applied for the determination of plaque stability. Real-time PCR was used for the detection of levels of lipid metabolism-related receptors. Electron microscopy and tandem fluorescent mRFP-GFP-LC3 plasmid was applied to test autophagy level. We found that β-arrestin-1 was highly increased in expression in plaque macrophage on the occurrence of atherosclerosis. Conditional myeloid knocking out β-arrestin-1 largely promotes plaque formation and vulnerability. In murine macrophage with lipid loading, knocking down β-arrestin-1 enhanced foam cell formation and levels of plasma and cellular cholesterol, while overexpressing β-arrestin-1 led to the opposite effects. The alleviative effects induced by macrophage β-arrestin-1 in atherosclerosis were involved in autophagy, based on the reduction of autophagy level with the knocking down of macrophage β-arrestin-1 and administration of autophagy inhibitors which largely attenuated the decreasing effect on foam cell formation. Our results demonstrated for the first time that macrophage β-arrestin-1 protected against atherosclerosis through the induction of autophagy.
Collapse
Affiliation(s)
- Bo-Zong Shao
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433 China
| | - Meng-Zhen Liu
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433 China
| | - Dan-Ni Zhu
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433 China
| | - Hui Yan
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433 China
| | - Ping Ke
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433 China
| | - Wei Wei
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Ting Han
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433 China
| | - Chong Liu
- Department of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai 200433 China.
| |
Collapse
|
41
|
Cismas S, Pasca S, Crudden C, Trocoli Drakensjo I, Suleymanova N, Zhang S, Gebhard B, Song D, Neo S, Shibano T, Smith TJ, Calin GA, Girnita A, Girnita L. Competing Engagement of β-arrestin Isoforms Balances IGF1R/p53 Signaling and Controls Melanoma Cell Chemotherapeutic Responsiveness. Mol Cancer Res 2023; 21:1288-1302. [PMID: 37584671 DOI: 10.1158/1541-7786.mcr-22-0871] [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: 11/06/2022] [Revised: 05/01/2023] [Accepted: 08/14/2023] [Indexed: 08/17/2023]
Abstract
Constraints on the p53 tumor suppressor pathway have long been associated with the progression, therapeutic resistance, and poor prognosis of melanoma, the most aggressive form of skin cancer. Likewise, the insulin-like growth factor type 1 receptor (IGF1R) is recognized as an essential coordinator of transformation, proliferation, survival, and migration of melanoma cells. Given that β-arrestin (β-arr) system critically governs the anti/pro-tumorigenic p53/IGF1R signaling pathways through their common E3 ubiquitin-protein ligase MDM2, we explore whether unbalancing this system downstream of IGF1R can enhance the response of melanoma cells to chemotherapy. Altering β-arr expression demonstrated that both β-arr1-silencing and β-arr2-overexpression (-β-arr1/+β-arr2) facilitated nuclear-to-cytosolic MDM2 translocation accompanied by decreased IGF1R expression, while increasing p53 levels, resulting in reduced cell proliferation/survival. Imbalance towards β-arr2 (-β-arr1/+β-arr2) synergizes with the chemotherapeutic agent, dacarbazine, in promoting melanoma cell toxicity. In both 3D spheroid models and in vivo in zebrafish models, this combination strategy, through dual IGF1R downregulation/p53 activation, limits melanoma cell growth, survival and metastatic spread. In clinical settings, analysis of the TCGA-SKCM patient cohort confirms β-arr1-/β-arr2+ imbalance as a metastatic melanoma vulnerability that may enhance therapeutic benefit. Our findings suggest that under steady-state conditions, IGF1R/p53-tumor promotion/suppression status-quo is preserved by β-arr1/2 homeostasis. Biasing this balance towards β-arr2 can limit the protumorigenic IGF1R activities while enhancing p53 activity, thus reducing multiple cancer-sustaining mechanisms. Combined with other therapeutics, this strategy improves patient responses and outcomes to therapies relying on p53 or IGF1R pathways. IMPLICATIONS Altogether, β-arrestin system bias downstream IGF1R is an important metastatic melanoma vulnerability that may be conductive for therapeutic benefit.
Collapse
Affiliation(s)
- Sonia Cismas
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Sylvya Pasca
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Caitrin Crudden
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Iara Trocoli Drakensjo
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Naida Suleymanova
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Simin Zhang
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Benjamin Gebhard
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Dawei Song
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Shiyong Neo
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Singapore Immunology Network SIgN, Agency for Science, Technology and Research, Singapore, Republic of Singapore
| | - Takashi Shibano
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Terry J Smith
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan
- Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - George A Calin
- Department of Experimental Therapeutics, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Ada Girnita
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Dermatology Department, Karolinska University Hospital, Stockholm, Sweden
| | - Leonard Girnita
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
42
|
Eissa AM, Hassanin MH, Ibrahim IAAEH. Hepatic β-arrestins: potential roles in liver health and disease. Mol Biol Rep 2023; 50:10399-10407. [PMID: 37843713 PMCID: PMC10676313 DOI: 10.1007/s11033-023-08898-0] [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: 05/29/2023] [Accepted: 10/04/2023] [Indexed: 10/17/2023]
Abstract
Β-arrestins are intracellular scaffolding proteins that have multifaceted roles in different types of disorders. In this review article, we gave a summary about the discovery, characterization and classification of these proteins and their intracellular functions. Moreover, this review article focused on the hepatic expression of β-arrestins and their hepatocellular distribution and function in each liver cell type. Also, we showed that β-arrestins are key regulators of distinct types of hepatic disorders. On the other hand, we addressed some important points that have never been studied before regarding the role of β-arrestins in certain types of hepatic disorders which needs more research efforts to cover.
Collapse
Affiliation(s)
| | | | - Islam A A E H Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
43
|
Zhou Y, Li F, Zou B, Zhou X, Luo L, Dong S, He Z, Zhang Z, Liao L, Liu H, Cai C, Gu D, Duan X. β-Arrestin2 promotes docetaxel resistance of castration-resistant prostate cancer via promoting hnRNP A1-mediated PKM2 alternative splicing. Discov Oncol 2023; 14:215. [PMID: 38019357 PMCID: PMC10686933 DOI: 10.1007/s12672-023-00740-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/26/2023] [Indexed: 11/30/2023] Open
Abstract
PURPOSE To investigate the influence of β-arrestin2 on the docetaxel resistance in castration-resistant prostate cancer (CRPC) and elucidate the underlying molecular mechanisms. METHODS PC3 and DU145 cells with stable β-arrestin2 overexpression and C4-2 cells with stable β-arrestin2 knockdown, were constructed via using lentivirus and puromycin selection. MTT and colony formation assays were carried out to investigate the effect of β-arrestin2 expression on the docetaxel resistance of CRPC cells. Glycolysis analysis was used to assess the glycolytic capacity modulated by β-arrestin2. GO enrichment analysis, gene set enrichment analysis and Spearman correlation test were carried out to explore the potential biological function and mechanism via using public data from GEO and TCGA. The expressions of PKM2, Phospho-PKM2, Phospho-ERK1/2 and hnRNP A1 were detected by western blot. Functional blocking experiments were carried out to confirm the roles of PKM2 and hnRNP A1 in the regulation of β-arrestin2's biological functions via silencing PKM2 or hnRNP A1 expression in cells with stable β-arrestin2 overexpression. Finally, nude mice xenograft models were established to confirm the experimental results of cell experiments. RESULTS β-Arrestin2 significantly decreased the sensitivity of CRPC cells to docetaxel stimulation, through enhancing the phosphorylation and expression of PKM2. Additionally, β-arrestin2 increased PKM2 phosphorylation via the ERK1/2 signaling pathway and induced PKM2 expression in a post-transcriptional manner through an hnRNP A1-dependent PKM alternative splicing mechanism, rather than by inhibiting its ubiquitination degradation. CONCLUSION Our findings indicate that the β-arrestin2/hnRNP A1/PKM2 pathway could be a promising target for treating docetaxel-resistant CRPC.
Collapse
Affiliation(s)
- Yuhao Zhou
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Kangda Road 1, Haizhu District, Guangzhou, 510230, Guangdong, China
| | - Fei Li
- Department of Pharmacy, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Bangyu Zou
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Kangda Road 1, Haizhu District, Guangzhou, 510230, Guangdong, China
| | - Xiaofeng Zhou
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Kangda Road 1, Haizhu District, Guangzhou, 510230, Guangdong, China
| | - Lianmin Luo
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Kangda Road 1, Haizhu District, Guangzhou, 510230, Guangdong, China
| | - Sicheng Dong
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Kangda Road 1, Haizhu District, Guangzhou, 510230, Guangdong, China
| | - Zhiqing He
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Kangda Road 1, Haizhu District, Guangzhou, 510230, Guangdong, China
| | - Zhixiong Zhang
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Kangda Road 1, Haizhu District, Guangzhou, 510230, Guangdong, China
| | - Liqiong Liao
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Kangda Road 1, Haizhu District, Guangzhou, 510230, Guangdong, China
| | - Hongxing Liu
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Kangda Road 1, Haizhu District, Guangzhou, 510230, Guangdong, China
| | - Chao Cai
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Kangda Road 1, Haizhu District, Guangzhou, 510230, Guangdong, China
| | - Di Gu
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Kangda Road 1, Haizhu District, Guangzhou, 510230, Guangdong, China.
| | - Xiaolu Duan
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou Institute of Urology, Kangda Road 1, Haizhu District, Guangzhou, 510230, Guangdong, China.
| |
Collapse
|
44
|
Liu H, Acharya S, Sudan SK, Hu L, Wu C, Cao Y, Li H, Zhang X. Comparative study of the molecular mechanisms underlying the G protein and β-arrestin-dependent pathways that lead to ERKs activation upon stimulation by dopamine D 2 receptor. FEBS J 2023; 290:5204-5233. [PMID: 37531324 DOI: 10.1111/febs.16921] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/09/2023] [Accepted: 08/01/2023] [Indexed: 08/04/2023]
Abstract
Dopamine D2 receptor (D2 R) has been shown to activate extracellular signal-regulated kinases (ERKs) via distinct pathways dependent on either G-protein or β-arrestin. However, there has not been a systematic study of the regulatory process of D2 R-mediated ERKs activation by G protein- versus β-arrestin-dependent signaling since D2 R stimulation of ERKs reflects the simultaneous action of both pathways. Here, we investigated that differential regulation of D2 R-mediated ERKs activation via these two pathways. Our results showed that G protein-dependent ERKs activation was transient, rapid, reached maximum level at around 2 min, and importantly, the activated ERKs were entirely confined to the cytoplasm. In contrast, β-arrestin-dependent ERKs activation was more sustained, slower, reached maximum level at around 10 min, and phosphorylated ERKs translocated into the nucleus. Src was found to be commonly involved in both the G protein- and β-arrestin-dependent pathway-mediated ERKs activation. Pertussis toxin Gi/o inhibitor, GRK2-CT, AG1478 epidermal growth factor receptor inhibitor, and wortmannin phosphoinositide 3-kinase inhibitor all blocked G protein-dependent ERKs activation. In contrast, GRK2 and β-Arr2 played a main role in β-arrestin-dependent ERKs activation. Receptor endocytosis showed minimal effect on the activation of ERKs mediated by both pathways. Furthermore, we found that the formation of a complex composed of phospho-ERKs, β-Arr2, and importinβ1 promoted the nuclear translocation of activated ERKs. The differential regulation of various cellular components, as well as temporal and spatial patterns of ERKs activation via these two pathways, suggest the existence of distinct physiological outcomes.
Collapse
Affiliation(s)
- Haiping Liu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| | - Srijan Acharya
- Mitchell Cancer Institute, School of Medicine, University of South Alabama, Mobile, AL, USA
| | - Sarabjeet Kour Sudan
- Mitchell Cancer Institute, School of Medicine, University of South Alabama, Mobile, AL, USA
| | - Li Hu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| | - Chengyan Wu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| | - Yongkai Cao
- Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, China
| | - Huijun Li
- Department of Pharmaceuticals, People's Hospital of Zunyi City Bo Zhou District, China
| | - Xiaohan Zhang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| |
Collapse
|
45
|
Eiger DS, Hicks C, Gardner J, Pham U, Rajagopal S. Location bias: A "Hidden Variable" in GPCR pharmacology. Bioessays 2023; 45:e2300123. [PMID: 37625014 PMCID: PMC11900906 DOI: 10.1002/bies.202300123] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/09/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023]
Abstract
G protein-coupled receptors (GPCRs) are the largest family of transmembrane receptors and primarily signal through two main effector proteins: G proteins and β-arrestins. Many agonists of GPCRs promote "biased" responses, in which different cellular signaling pathways are activated with varying efficacies. The mechanisms underlying biased signaling have not been fully elucidated, with many potential "hidden variables" that regulate this behavior. One contributor is "location bias," which refers to the generation of unique signaling cascades from a given GPCR depending upon the cellular location at which the receptor is signaling. Here, we review evidence that GPCRs are expressed at and traffic to various subcellular locations and discuss how location bias can impact the pharmacologic properties and characterization of GPCR agonists. We also evaluate how differences in subcellular environments can modulate GPCR signaling, highlight the physiological significance of subcellular GPCR signaling, and discuss the therapeutic potential of exploiting GPCR location bias.
Collapse
Affiliation(s)
- Dylan Scott Eiger
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, 02215, USA
| | - Chloe Hicks
- Trinity College, Duke University, Durham, NC, 27710, USA
| | - Julia Gardner
- Trinity College, Duke University, Durham, NC, 27710, USA
| | - Uyen Pham
- Department of Biochemistry, Duke University, Durham, NC, 27710, USA
| | - Sudarshan Rajagopal
- Department of Biochemistry, Duke University, Durham, NC, 27710, USA
- Department of Medicine, Duke University, Durham, NC, 27710, USA
| |
Collapse
|
46
|
Burghi V, Paradis JS, Officer A, Adame-Garcia SR, Wu X, Matthees ESF, Barsi-Rhyne B, Ramms DJ, Clubb L, Acosta M, Tamayo P, Bouvier M, Inoue A, von Zastrow M, Hoffmann C, Gutkind JS. Gαs is dispensable for β-arrestin coupling but dictates GRK selectivity and is predominant for gene expression regulation by β2-adrenergic receptor. J Biol Chem 2023; 299:105293. [PMID: 37774973 PMCID: PMC10641165 DOI: 10.1016/j.jbc.2023.105293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 09/03/2023] [Accepted: 09/14/2023] [Indexed: 10/01/2023] Open
Abstract
β-arrestins play a key role in G protein-coupled receptor (GPCR) internalization, trafficking, and signaling. Whether β-arrestins act independently of G protein-mediated signaling has not been fully elucidated. Studies using genome-editing approaches revealed that whereas G proteins are essential for mitogen-activated protein kinase activation by GPCRs., β-arrestins play a more prominent role in signal compartmentalization. However, in the absence of G proteins, GPCRs may not activate β-arrestins, thereby limiting the ability to distinguish G protein from β-arrestin-mediated signaling events. We used β2-adrenergic receptor (β2AR) and its β2AR-C tail mutant expressed in human embryonic kidney 293 cells wildtype or CRISPR-Cas9 gene edited for Gαs, β-arrestin1/2, or GPCR kinases 2/3/5/6 in combination with arrestin conformational sensors to elucidate the interplay between Gαs and β-arrestins in controlling gene expression. We found that Gαs is not required for β2AR and β-arrestin conformational changes, β-arrestin recruitment, and receptor internalization, but that Gαs dictates the GPCR kinase isoforms involved in β-arrestin recruitment. By RNA-Seq analysis, we found that protein kinase A and mitogen-activated protein kinase gene signatures were activated by stimulation of β2AR in wildtype and β-arrestin1/2-KO cells but absent in Gαs-KO cells. These results were validated by re-expressing Gαs in the corresponding KO cells and silencing β-arrestins in wildtype cells. These findings were extended to cellular systems expressing endogenous levels of β2AR. Overall, our results support that Gs is essential for β2AR-promoted protein kinase A and mitogen-activated protein kinase gene expression signatures, whereas β-arrestins initiate signaling events modulating Gαs-driven nuclear transcriptional activity.
Collapse
Affiliation(s)
- Valeria Burghi
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Justine S Paradis
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Adam Officer
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Sendi Rafael Adame-Garcia
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Xingyu Wu
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Edda S F Matthees
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - Benjamin Barsi-Rhyne
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - Dana J Ramms
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Lauren Clubb
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Monica Acosta
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA
| | - Pablo Tamayo
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA
| | - Michel Bouvier
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Québec, Canada
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Mark von Zastrow
- Department of Psychiatry and Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California, USA
| | - Carsten Hoffmann
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Jena, Germany
| | - J Silvio Gutkind
- Moores Cancer Center, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA.
| |
Collapse
|
47
|
Rahman RJ, Rijal R, Jing S, Chen TA, Ismail I, Gomer RH. Polyphosphate uses mTOR, pyrophosphate, and Rho GTPase components to potentiate bacterial survival in Dictyostelium. mBio 2023; 14:e0193923. [PMID: 37754562 PMCID: PMC10653871 DOI: 10.1128/mbio.01939-23] [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: 07/25/2023] [Accepted: 07/31/2023] [Indexed: 09/28/2023] Open
Abstract
IMPORTANCE Although most bacteria are quickly killed after phagocytosis by a eukaryotic cell, some pathogenic bacteria escape death after phagocytosis. Pathogenic Mycobacterium species secrete polyP, and the polyP is necessary for the bacteria to prevent their killing after phagocytosis. Conversely, exogenous polyP prevents the killing of ingested bacteria that are normally killed after phagocytosis by human macrophages and the eukaryotic microbe Dictyostelium discoideum. This suggests the possibility that in these cells, a signal transduction pathway is used to sense polyP and prevent killing of ingested bacteria. In this report, we identify key components of the polyP signal transduction pathway in D. discoideum. In cells lacking these components, polyP is unable to inhibit killing of ingested bacteria. The pathway components have orthologs in human cells, and an exciting possibility is that pharmacologically blocking this pathway in human macrophages would cause them to kill ingested pathogens such as Mycobacterium tuberculosis.
Collapse
Affiliation(s)
- Ryan J. Rahman
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Ramesh Rijal
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Shiyu Jing
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Te-An Chen
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Issam Ismail
- Department of Biology, Texas A&M University, College Station, Texas, USA
| | - Richard H. Gomer
- Department of Biology, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
48
|
Li Y, Li K, Pan T, Xie Q, Cheng Y, Wu X, Xu R, Liu X, Liu L, Gao J, Yuan W, Qu X, Cui S. Translocation of IGF-1R in endoplasmic reticulum enhances SERCA2 activity to trigger Ca 2+ER perturbation in hepatocellular carcinoma. Acta Pharm Sin B 2023; 13:3744-3755. [PMID: 37719369 PMCID: PMC10501870 DOI: 10.1016/j.apsb.2023.05.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 09/19/2023] Open
Abstract
The well-known insulin-like growth factor 1 (IGF1)/IGF-1 receptor (IGF-1R) signaling pathway is overexpressed in many tumors, and is thus an attractive target for cancer treatment. However, results have often been disappointing due to crosstalk with other signals. Here, we report that IGF-1R signaling stimulates the growth of hepatocellular carcinoma (HCC) cells through the translocation of IGF-1R into the ER to enhance sarco-endoplasmic reticulum calcium ATPase 2 (SERCA2) activity. In response to ligand binding, IGF-1Rβ is translocated into the ER by β-arrestin2 (β-arr2). Mass spectrometry analysis identified SERCA2 as a target of ER IGF-1Rβ. SERCA2 activity is heavily dependent on the increase in ER IGF-1Rβ levels. ER IGF-1Rβ phosphorylates SERCA2 on Tyr990 to enhance its activity. Mutation of SERCA2-Tyr990 disrupted the interaction of ER IGF-1Rβ with SERCA2, and therefore ER IGF-1Rβ failed to promote SERCA2 activity. The enhancement of SERCA2 activity triggered Ca2+ER perturbation, leading to an increase in autophagy. Thapsigargin blocked the interaction between SERCA2 and ER IGF-1Rβ and therefore SERCA2 activity, resulting in inhibition of HCC growth. In conclusion, the translocation of IGF-1R into the ER triggers Ca2+ER perturbation by enhancing SERCA2 activity through phosphorylating Tyr990 in HCC.
Collapse
Affiliation(s)
- Yanan Li
- Department of Toxicology and Sanitary Chemistry, Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Keqin Li
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Ting Pan
- Department of Toxicology and Sanitary Chemistry, Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Qiaobo Xie
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yuyao Cheng
- Department of Toxicology and Sanitary Chemistry, Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Xinfeng Wu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Rui Xu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xiaohui Liu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Li Liu
- Department of Pharmacology, Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Jiangming Gao
- Department of Pharmacology, Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Wenmin Yuan
- Department of Pharmacology, Marine Biomedical Research Institute of Qingdao, Qingdao 266071, China
| | - Xianjun Qu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Shuxiang Cui
- Department of Toxicology and Sanitary Chemistry, Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, China
| |
Collapse
|
49
|
Ramos‐Gonzalez N, Groom S, Sutcliffe KJ, Bancroft S, Bailey CP, Sessions RB, Henderson G, Kelly E. Carfentanil is a β-arrestin-biased agonist at the μ opioid receptor. Br J Pharmacol 2023; 180:2341-2360. [PMID: 37005796 PMCID: PMC10952505 DOI: 10.1111/bph.16084] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 03/10/2023] [Accepted: 03/28/2023] [Indexed: 04/04/2023] Open
Abstract
BACKGROUND AND PURPOSE The illicit use of fentanyl-like drugs (fentanyls), which are μ opioid receptor agonists, and the many overdose deaths that result, has become a major problem. Fentanyls are very potent in vivo, leading to respiratory depression and death. However, the efficacy and possible signalling bias of different fentanyls is not clearly known. Here, we compared the relative efficacy and bias of a series of fentanyls. EXPERIMENTAL APPROACH For agonist signalling bias and efficacy measurements, Bioluminescence Resonance Energy Transfer experiments were undertaken in HEK293T cells transiently transfected with μ opioid receptors, to assess Gi protein activation and β-arrestin 2 recruitment. Agonist-induced cell surface receptor loss was assessed using an enzyme-linked immunosorbent assay, whilst agonist-induced G protein-coupled inwardly rectifying potassium channel current activation was measured electrophysiologically from rat locus coeruleus slices. Ligand poses in the μ opioid receptor were determined in silico using molecular dynamics simulations. KEY RESULTS Relative to the reference ligand DAMGO, carfentanil was β-arrestin-biased, whereas fentanyl, sufentanil and alfentanil did not display bias. Carfentanil induced potent and extensive cell surface receptor loss, whilst the marked desensitisation of G protein-coupled inwardly rectifying potassium channel currents in the continued presence of carfentanil in neurones was prevented by a GRK2/3 inhibitor. Molecular dynamics simulations suggested unique interactions of carfentanil with the orthosteric site of the receptor that could underlie the bias. CONCLUSIONS AND IMPLICATIONS Carfentanil is a β-arrestin-biased opioid drug at the μ receptor. It is uncertain how such bias influences in vivo effects of carfentanil relative to other fentanyls.
Collapse
Affiliation(s)
| | - Sam Groom
- Department of Pharmacy and PharmacologyUniversity of BathBathUK
| | - Katy J. Sutcliffe
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Sukhvinder Bancroft
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Chris P. Bailey
- Department of Pharmacy and PharmacologyUniversity of BathBathUK
| | | | - Graeme Henderson
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| | - Eamonn Kelly
- School of Physiology, Pharmacology and NeuroscienceUniversity of BristolBristolUK
| |
Collapse
|
50
|
Birch CA, Wedegaertner H, Orduña-Castillo LB, Gonzalez Ramirez ML, Qin H, Trejo J. Endothelial APC/PAR1 distinctly regulates cytokine-induced pro-inflammatory VCAM-1 expression. Front Mol Biosci 2023; 10:1211597. [PMID: 37692066 PMCID: PMC10483999 DOI: 10.3389/fmolb.2023.1211597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/04/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction: Dysfunction of the endothelium impairs its' protective role and promotes inflammation and progression of vascular diseases. Activated Protein C (APC) elicits endothelial cytoprotective responses including barrier stabilization, anti-inflammatory and anti-apoptotic responses through the activation of the G protein-coupled receptor (GPCR) protease-activated receptor-1 (PAR1) and is a promising therapeutic. Despite recent advancements in developing new Activated protein C variants with clinical potential, the mechanism by which APC/PAR1 promotes different cytoprotective responses remains unclear and is important to understand to advance Activated protein C and new targets as future therapeutics. Here we examined the mechanisms by which APC/PAR1 attenuates cytokine-induced pro-inflammatory vascular cell adhesion molecule (VCAM-1) expression, a key mediator of endothelial inflammatory responses. Methods: Quantitative multiplexed mass spectrometry analysis of Activated protein C treated endothelial cells, endothelial cell transcriptomics database (EndoDB) online repository queries, biochemical measurements of protein expression, quantitative real-time polymerase chain reaction (RT-qPCR) measurement of mRNA transcript abundance, pharmacological inhibitors and siRNA transfections of human cultured endothelial cells. Results: Here we report that Activated Protein C modulates phosphorylation of tumor necrosis factor (TNF)-α signaling pathway components and attenuates of TNF-α induced VCAM-1 expression independent of mRNA stability. Unexpectedly, we found a critical role for the G protein-coupled receptor co-receptor sphingosine-1 phosphate receptor-1 (S1PR1) and the G protein receptor kinase-2 (GRK2) in mediating APC/PAR1 anti-inflammatory responses in endothelial cells. Discussion: This study provides new knowledge of the mechanisms by which different APC/PAR1 cytoprotective responses are mediated through discrete β-arrestin-2-driven signaling pathways modulated by specific G protein-coupled receptor co-receptors and GRKs.
Collapse
Affiliation(s)
- Cierra A. Birch
- Department of Pharmacology, School of Medicine, University of California, San Diego, CA, United States
| | - Helen Wedegaertner
- Department of Pharmacology, School of Medicine, University of California, San Diego, CA, United States
- Biomedical Sciences Graduate Program, University of California, San Diego, CA, United States
| | - Lennis B. Orduña-Castillo
- Department of Pharmacology, School of Medicine, University of California, San Diego, CA, United States
| | | | - Huaping Qin
- Department of Pharmacology, School of Medicine, University of California, San Diego, CA, United States
| | - JoAnn Trejo
- Department of Pharmacology, School of Medicine, University of California, San Diego, CA, United States
| |
Collapse
|