1
|
Zhang H, Wu T, Wu Y, Peng Y, Wei X, Lu T, Jiao Y. Binding sites and design strategies for small molecule GLP-1R agonists. Eur J Med Chem 2024; 275:116632. [PMID: 38959726 DOI: 10.1016/j.ejmech.2024.116632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024]
Abstract
Glucagon-like peptide-1 receptor (GLP-1R) is a pivotal receptor involved in blood glucose regulation and influencing feeding behavior. It has received significant attention in the treatment of obesity and diabetes due to its potent incretin effect. Peptide GLP-1 receptor agonists (GLP-1RAs) have achieved tremendous success in the market, driving the vigorous development of small molecule GLP-1RAs. Currently, several small molecules have entered the clinical research stage. Additionally, recent discoveries of GLP-1R positive allosteric modulators (PAMs) are also unveiling new regulatory patterns and treatment methods. This article reviews the structure and functional mechanisms of GLP-1R, recent reports on small molecule GLP-1RAs and PAMs, as well as the optimization process. Furthermore, it combines computer simulations to analyze structure-activity relationships (SAR) studies, providing a foundation for exploring new strategies for designing small molecule GLP-1RAs.
Collapse
Affiliation(s)
- Haibo Zhang
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China
| | - Tianxiao Wu
- Jiangsu Vcare PharmaTech Co., Ltd., 136 Huakang Road, Nanjing, 211800, China
| | - Yong Wu
- Jiangsu Vcare PharmaTech Co., Ltd., 136 Huakang Road, Nanjing, 211800, China
| | - Yuran Peng
- Jiangsu Vcare PharmaTech Co., Ltd., 136 Huakang Road, Nanjing, 211800, China
| | - Xian Wei
- Department of Pharmacy, Youjiang Medical University for Nationalities, 98 ChengXiang Road, Baise, 533000, China.
| | - Tao Lu
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China.
| | - Yu Jiao
- School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, 211198, China.
| |
Collapse
|
2
|
Gareri C, Pfeiffer CT, Jiang X, Paulo JA, Gygi SP, Pham U, Chundi A, Wingler LM, Staus DP, Stepniewski TM, Selent J, Lucero EY, Grogan A, Rajagopal S, Rockman HA. Phosphorylation patterns in the AT1R C-terminal tail specify distinct downstream signaling pathways. Sci Signal 2024; 17:eadk5736. [PMID: 39137246 PMCID: PMC11443182 DOI: 10.1126/scisignal.adk5736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/01/2023] [Accepted: 07/23/2024] [Indexed: 08/15/2024]
Abstract
Different ligands stabilize specific conformations of the angiotensin II type 1 receptor (AT1R) that direct distinct signaling cascades mediated by heterotrimeric G proteins or β-arrestin. These different active conformations are thought to engage distinct intracellular transducers because of differential phosphorylation patterns in the receptor C-terminal tail (the "barcode" hypothesis). Here, we identified the AT1R barcodes for the endogenous agonist AngII, which stimulates both G protein activation and β-arrestin recruitment, and for a synthetic biased agonist that only stimulates β-arrestin recruitment. The endogenous and β-arrestin-biased agonists induced two different ensembles of phosphorylation sites along the C-terminal tail. The phosphorylation of eight serine and threonine residues in the proximal and middle portions of the tail was required for full β-arrestin functionality, whereas phosphorylation of the serine and threonine residues in the distal portion of the tail had little influence on β-arrestin function. Similarly, molecular dynamics simulations showed that the proximal and middle clusters of phosphorylated residues were critical for stable β-arrestin-receptor interactions. These findings demonstrate that ligands that stabilize different receptor conformations induce different phosphorylation clusters in the C-terminal tail as barcodes to evoke distinct receptor-transducer engagement, receptor trafficking, and signaling.
Collapse
Affiliation(s)
- Clarice Gareri
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Conrad T. Pfeiffer
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Xue Jiang
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Uyen Pham
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
| | - Anand Chundi
- Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA
| | - Laura M. Wingler
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | - Dean P. Staus
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Tomasz Maciej Stepniewski
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM) - Pompeu Fabra University (UPF), 08003 Barcelona, Spain
- Faculty of Chemistry, Biological and Chemical Research Center, University of Warsaw, Warsaw, Poland
- InterAx Biotech AG, PARK InnovAARE, 5234 Villigen, Switzerland
| | - Jana Selent
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM) - Pompeu Fabra University (UPF), 08003 Barcelona, Spain
| | - Emilio Y. Lucero
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Alyssa Grogan
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Sudarshan Rajagopal
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
| | - Howard A. Rockman
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
3
|
Blum NK, Schaffner A, Drube J, Nagel F, Reinscheid RK, Hoffmann C, Schulz S. Rapid elucidation of agonist-driven regulation of the neurokinin 1 receptor using a GPCR phosphorylation immunoassay. Eur J Pharmacol 2024; 973:176587. [PMID: 38642667 DOI: 10.1016/j.ejphar.2024.176587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/22/2024]
Abstract
Agonist-induced phosphorylation is a crucial step in the activation/deactivation cycle of G protein-coupled receptors (GPCRs), but direct determination of individual phosphorylation events has remained a major challenge. We have recently developed a bead-based immunoassay for the quantitative assessment of agonist-induced GPCR phosphorylation that can be performed entirely in 96-well plates, thus eliminating the need for western blot analysis. In the present study, we adapted this assay to three novel phosphosite-specific antibodies directed against the neurokinin 1 (NK1) receptor, namely pS338/pT339-NK1, pT344/pS347-NK1, and pT356/pT357-NK1. We found that substance P (SP) stimulated concentration-dependent phosphorylation of all three sites, which could be completely blocked in the presence of the NK1 receptor antagonist aprepitant. The other two endogenous ligands of the tachykinin family, neurokinin A (NKA) and neurokinin B (NKB), were also able to induce NK1 receptor phosphorylation, but to a much lesser extent than substance P. Interestingly, substance P promoted phosphorylation of the two distal sites more efficiently than that of the proximal site. The proximal site was identified as a substrate for phosphorylation by protein kinase C. Analysis of GPCR kinase (GRK)-knockout cells revealed that phosphorylation was mediated by all four GRK isoforms to similar extents at the T344/S347 and the T356/T357 cluster. Knockout of all GRKs resulted in abolition of all phosphorylation signals highlighting the importance of these kinases in agonist-mediated receptor phosphorylation. Thus, the 7TM phosphorylation assay technology allows for rapid and detailed analyses of GPCR phosphorylation.
Collapse
Affiliation(s)
- Nina K Blum
- Institut für Pharmakologie und Toxikologie, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Drackendorfer Str. 1, D-07747, Jena, Germany
| | - Anne Schaffner
- Institut für Pharmakologie und Toxikologie, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Drackendorfer Str. 1, D-07747, Jena, Germany
| | - Julia Drube
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Str. 2, D-07745, Jena, Germany
| | - Falko Nagel
- 7TM Antibodies GmbH, Hans-Knöll-Str. 6, D-07745, Jena, Germany
| | - Rainer K Reinscheid
- Institut für Pharmakologie und Toxikologie, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Drackendorfer Str. 1, D-07747, Jena, Germany
| | - Carsten Hoffmann
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Str. 2, D-07745, Jena, Germany
| | - Stefan Schulz
- Institut für Pharmakologie und Toxikologie, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Drackendorfer Str. 1, D-07747, Jena, Germany; 7TM Antibodies GmbH, Hans-Knöll-Str. 6, D-07745, Jena, Germany.
| |
Collapse
|
4
|
Guven B, Onay-Besikci A. Past and present of beta arrestins: A new perspective on insulin secretion and effect. Eur J Pharmacol 2023; 956:175952. [PMID: 37541367 DOI: 10.1016/j.ejphar.2023.175952] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/24/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023]
Abstract
BACKGROUND Beta arrestins had been known as intracellular adaptors that uncouple and inactivate the G protein-coupled receptors that they interact with. Their roles as signal initiators for some receptors have recently been recognized. SCOPE OF REVIEW In this review, we focused on their role in mediating metabolic modulation primarily in relation to insulin signaling. Commenced by the upstream receptor, they seem to act like intracellular hubs that divert the metabolic profile of the cell. The amount of metabolic substrates in circulation and their usage/deposition by tissues are controlled by the contribution of all systems in the organism. This control is enabled by the release of hormones such as insulin, glucagon and glucagon-like peptide-1. Intriguingly, some ligands -either agonists or antagonists-of different classes of receptors have preferential properties mediated by β arrestins. This is not surprizing considering that substrate supply and usage should parallel physiological function such as hormone release or muscle contraction. MAJOR CONCLUSIONS Available data indicate that β arrestins conduct the regulatory role in insulin secretion and action. They may be good candidates to target when the upstream signal demands the function that may compromise the cell. An example is carvedilol that is protective by preventing the stimulatory effects of excessive catecholamines, stimulates mitochondrial function and has preferential clinical outcomes in metabolic disorders.
Collapse
Affiliation(s)
- Berna Guven
- Faculty of Pharmacy, Department of Pharmacology, Ankara University, Ankara, Turkey
| | - Arzu Onay-Besikci
- Faculty of Pharmacy, Department of Pharmacology, Ankara University, Ankara, Turkey.
| |
Collapse
|
5
|
Liu H, Ma H, Zeng X, Wu C, Acharya S, Sudan SK, Zhang X. Ubiquitination of GRK2 Is Required for the β-Arrestin-Biased Signaling Pathway of Dopamine D2 Receptors to Activate ERK Kinases. Int J Mol Sci 2023; 24:10031. [PMID: 37373182 DOI: 10.3390/ijms241210031] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
A class-A GPCR dopamine D2 receptor (D2R) plays a critical role in the proper functioning of neuronal circuits through the downstream activation of both G-protein- and β-arrestin-dependent signaling pathways. Understanding the signaling pathways downstream of D2R is critical for developing effective therapies with which to treat dopamine (DA)-related disorders such as Parkinson's disease and schizophrenia. Extensive studies have focused on the regulation of D2R-mediated extracellular-signal-regulated kinase (ERK) 1/2 signaling; however, the manner in which ERKs are activated upon the stimulation of a specific signaling pathway of D2R remains unclear. The present study conducted a variety of experimental techniques, including loss-of-function experiments, site-directed mutagenesis, and the determination of protein interactions, in order to investigate the mechanisms underlying β-arrestin-biased signaling-pathway-mediated ERK activation. We found that the stimulation of the D2R β-arrestin signaling pathway caused Mdm2, an E3 ubiquitin ligase, to move from the nucleus to the cytoplasm and interact with tyrosine phosphorylated G-protein-coupled receptor kinase 2 (GRK2), which was facilitated by Src, a non-receptor tyrosine kinase. This interaction led to the ubiquitination of GRK2, which then moved to the plasma membrane and interacted with activated D2R, followed by the phosphorylation of D2R as well as the mediation of ERK activation. In conclusion, Mdm2-mediated GRK2 ubiquitination, which is selectively triggered by the stimulation of the D2R β-arrestin signaling pathway, is necessary for GRK2 membrane translocation and its interaction with D2R, which in turn mediates downstream ERK signaling. This study is primarily novel and provides essential information with which to better understand the detailed mechanisms of D2R-dependent signaling.
Collapse
Affiliation(s)
- Haiping Liu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Haixiang Ma
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Xingyue Zeng
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Chengyan Wu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| | - Srijan Acharya
- Mitchell Cancer Institute, School of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Sarabjeet Kour Sudan
- Mitchell Cancer Institute, School of Medicine, University of South Alabama, Mobile, AL 36604, USA
| | - Xiaohan Zhang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China
| |
Collapse
|
6
|
Wang D, Yao Y, Wang S, Hou Y, Zhao L, Wang H, Chen H, Xu J. Structural Insights into M1 Muscarinic Acetylcholine Receptor Signaling Bias between Gαq and β-Arrestin through BRET Assays and Molecular Docking. Int J Mol Sci 2023; 24:ijms24087356. [PMID: 37108518 PMCID: PMC10138654 DOI: 10.3390/ijms24087356] [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: 03/08/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
The selectivity of drugs for G protein-coupled receptor (GPCR) signaling pathways is crucial for their therapeutic efficacy. Different agonists can cause receptors to recruit effector proteins at varying levels, thus inducing different signaling responses, called signaling bias. Although several GPCR-biased drugs are currently being developed, only a limited number of biased ligands have been identified regarding their signaling bias for the M1 muscarinic acetylcholine receptor (M1mAChR), and the mechanism is not yet well understood. In this study, we utilized bioluminescence resonance energy transfer (BRET) assays to compare the efficacy of six agonists in inducing Gαq and β-arrestin2 binding to M1mAChR. Our findings reveal notable variations in agonist efficacy in the recruitment of Gαq and β-arrestin2. Pilocarpine preferentially promoted the recruitment of β-arrestin2 (∆∆RAi = -0.5), while McN-A-343 (∆∆RAi = 1.5), Xanomeline (∆∆RAi = 0.6), and Iperoxo (∆∆RAi = 0.3) exhibited a preference for the recruitment of Gαq. We also used commercial methods to verify the agonists and obtained consistent results. Molecular docking revealed that certain residues (e.g., Y404, located in TM7 of M1mAChR) could play crucial roles in Gαq signaling bias by interacting with McN-A-343, Xanomeline, and Iperoxo, whereas other residues (e.g., W378 and Y381, located in TM6) contributed to β-arrestin recruitment by interacting with Pilocarpine. The preference of activated M1mAChR for different effectors may be due to significant conformational changes induced by biased agonists. By characterizing bias towards Gαq and β-arrestin2 recruitment, our study provides insights into M1mAChR signaling bias.
Collapse
Affiliation(s)
- Dongxue Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yunjin Yao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shiqi Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yifei Hou
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lanxue Zhao
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hao Wang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hongzhuan Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianrong Xu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
7
|
Fritzwanker S, Nagel F, Kliewer A, Stammer V, Schulz S. In situ visualization of opioid and cannabinoid drug effects using phosphosite-specific GPCR antibodies. Commun Biol 2023; 6:419. [PMID: 37061609 PMCID: PMC10105690 DOI: 10.1038/s42003-023-04786-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 03/30/2023] [Indexed: 04/17/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are important signal transducers that are phosphorylated upon activation at intracellular serine and threonine residues. Although antibodies that specifically recognize the phosphorylation state of GPCRs have been available for many years, efficient immunolocalization of phosphorylated receptors in their tissues of origin has not been possible. Here, we show that phosphorylation of receptors is highly unstable during routine immunohistochemical procedures, requiring the use of appropriate phosphatase inhibitors particular during tissue perfusion, post-fixation, and cryoprotection but not during immunostaining of tissue sections. We provide proof of concept using phosphorylation state-specific μ-opioid receptor (MOP) and cannabinoid receptor 1 (CB1) antibodies. Indeed, three of four well-characterized phosphosite-specific MOP antibodies, including pS375-MOP, pT376-MOP, and pT379-MOP, showed robust neuronal immunostaining in brain and spinal cord sections of opioid-treated mice only after inclusion of phosphatase inhibitors. We then extended this approach to the CB1 receptor and demonstrated that one of three newly-generated phosphosite-specific CB1 antibodies, namely pS425-CB1, showed striking staining of fibers and varicosities in brain slices from cannabinoid-treated mice. Although subsequent experiments showed that phospho-CB1 immunostaining was less sensitive to phosphatases, we conclude that the use of phosphatase inhibitors should always be considered in the development of immunohistochemical procedures for new phosphosite-specific GPCR antibodies. In summary, we anticipate that this improved protocol will facilitate the widespread use of phosphorylation state-specific antibodies to monitor the activation of endogenous GPCRs under physiological and pharmacological conditions. Our approach may also prove useful to confirm target engagement of GPCR drug candidates in native tissues.
Collapse
Affiliation(s)
- Sebastian Fritzwanker
- Institut für Pharmakologie und Toxikologie, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Drackendorfer Straße 1, D-07747, Jena, Germany
| | - Falko Nagel
- 7TM Antibodies GmbH, Hans-Knöll-Straße 6, D-07745, Jena, Germany
| | - Andrea Kliewer
- Institut für Pharmakologie und Toxikologie, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Drackendorfer Straße 1, D-07747, Jena, Germany
| | - Viviane Stammer
- Institut für Pharmakologie und Toxikologie, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Drackendorfer Straße 1, D-07747, Jena, Germany
| | - Stefan Schulz
- Institut für Pharmakologie und Toxikologie, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Drackendorfer Straße 1, D-07747, Jena, Germany.
- 7TM Antibodies GmbH, Hans-Knöll-Straße 6, D-07745, Jena, Germany.
| |
Collapse
|
8
|
Kaufmann J, Blum NK, Nagel F, Schuler A, Drube J, Degenhart C, Engel J, Eickhoff JE, Dasgupta P, Fritzwanker S, Guastadisegni M, Schulte C, Miess-Tanneberg E, Maric HM, Spetea M, Kliewer A, Baumann M, Klebl B, Reinscheid RK, Hoffmann C, Schulz S. A bead-based GPCR phosphorylation immunoassay for high-throughput ligand profiling and GRK inhibitor screening. Commun Biol 2022; 5:1206. [PMID: 36352263 PMCID: PMC9646841 DOI: 10.1038/s42003-022-04135-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 10/19/2022] [Indexed: 11/10/2022] Open
Abstract
Analysis of agonist-driven phosphorylation of G protein-coupled receptors (GPCRs) can provide valuable insights into the receptor activation state and ligand pharmacology. However, to date, assessment of GPCR phosphorylation using high-throughput applications has been challenging. We have developed and validated a bead-based immunoassay for the quantitative assessment of agonist-induced GPCR phosphorylation that can be performed entirely in multiwell cell culture plates. The assay involves immunoprecipitation of affinity-tagged receptors using magnetic beads followed by protein detection using phosphorylation state-specific and phosphorylation state-independent anti-GPCR antibodies. As proof of concept, five prototypical GPCRs (MOP, C5a1, D1, SST2, CB2) were treated with different agonizts and antagonists, and concentration-response curves were generated. We then extended our approach to establish selective cellular GPCR kinase (GRK) inhibitor assays, which led to the rapid identification of a selective GRK5/6 inhibitor (LDC8988) and a highly potent pan-GRK inhibitor (LDC9728). In conclusion, this versatile GPCR phosphorylation assay can be used extensively for ligand profiling and inhibitor screening. A G protein-coupled receptors (GPCRs) phosphorylation assay for cell culture plates can be used for ligand profiling and inhibitor screening, as evidenced by the identification of two GRK inhibitor compounds.
Collapse
|
9
|
Fonseca PAS, Alonso-García M, Pelayo R, Marina H, Esteban-Blanco C, Mateo J, Gutiérrez-Gil B, Arranz JJ, Suárez-Vega A. Integrated analyses of the methylome and transcriptome to unravel sex differences in the perirenal fat from suckling lambs. Front Genet 2022; 13:1035063. [PMID: 36386829 PMCID: PMC9663842 DOI: 10.3389/fgene.2022.1035063] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/18/2022] [Indexed: 11/25/2022] Open
Abstract
In sheep, differences were observed regarding fat accumulation and fatty acid (FA) composition between males and females, which may impact the quality and organoleptic characteristics of the meat. The integration of different omics technologies is a relevant approach for investigating biological and genetic mechanisms associated with complex traits. Here, the perirenal tissue of six male and six female Assaf suckling lambs was evaluated using RNA sequencing and whole-genome bisulfite sequencing (WGBS). A multiomic discriminant analysis using multiblock (s)PLS-DA allowed the identification of 314 genes and 627 differentially methylated regions (within these genes), which perfectly discriminate between males and females. These candidate genes overlapped with previously reported QTLs for carcass fat volume and percentage of different FAs in milk and meat from sheep. Additionally, differentially coexpressed (DcoExp) modules of genes between males (nine) and females (three) were identified that harbour 22 of these selected genes. Interestingly, these DcoExp were significantly correlated with fat percentage in different deposits (renal, pelvic, subcutaneous and intramuscular) and were associated with relevant biological processes for adipogenesis, adipocyte differentiation, fat volume and FA composition. Consequently, these genes may potentially impact adiposity and meat quality traits in a sex-specific manner, such as juiciness, tenderness and flavour.
Collapse
Affiliation(s)
- Pablo A. S. Fonseca
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, León, Spain
| | - María Alonso-García
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, León, Spain
| | - Rocio Pelayo
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, León, Spain
| | - Hector Marina
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, León, Spain
| | - Cristina Esteban-Blanco
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, León, Spain
| | - Javier Mateo
- Departamento de Higiene y Tecnología de Los Alimentos, Facultad de Veterinaria, Universidad de León, León, Spain
| | - Beatriz Gutiérrez-Gil
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, León, Spain
| | - Juan-José Arranz
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, León, Spain,*Correspondence: Juan-José Arranz,
| | - Aroa Suárez-Vega
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, León, Spain
| |
Collapse
|
10
|
Wu Y, Wang S, Wang H, Hu B, Wang J. Selectivity mechanism of GRK2/5 inhibition through in silico investigation. Comput Biol Chem 2022; 101:107786. [DOI: 10.1016/j.compbiolchem.2022.107786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/02/2022] [Accepted: 11/05/2022] [Indexed: 11/09/2022]
|
11
|
Martin L, Ibrahim M, Gomez K, Yu J, Cai S, Chew LA, Bellampalli SS, Moutal A, Largent-Milnes T, Porreca F, Khanna R, Olivera BM, Patwardhan A. Conotoxin contulakin-G engages a neurotensin receptor 2/R-type calcium channel (Cav2.3) pathway to mediate spinal antinociception. Pain 2022; 163:1751-1762. [PMID: 35050960 PMCID: PMC9198109 DOI: 10.1097/j.pain.0000000000002561] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 12/07/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT Intrathecal application of contulakin-G (CGX), a conotoxin peptide and a neurotensin analogue, has been demonstrated to be safe and potentially analgesic in humans. However, the mechanism of action for CGX analgesia is unknown. We hypothesized that spinal application of CGX produces antinociception through activation of the presynaptic neurotensin receptor (NTSR)2. In this study, we assessed the mechanisms of CGX antinociception in rodent models of inflammatory and neuropathic pain. Intrathecal administration of CGX, dose dependently, inhibited thermal and mechanical hypersensitivities in rodents of both sexes. Pharmacological and clustered regularly interspaced short palindromic repeats/Cas9 editing of NTSR2 reversed CGX-induced antinociception without affecting morphine analgesia. Electrophysiological and gene editing approaches demonstrated that CGX inhibition was dependent on the R-type voltage-gated calcium channel (Cav2.3) in sensory neurons. Anatomical studies demonstrated coexpression of NTSR2 and Cav2.3 in dorsal root ganglion neurons. Finally, synaptic fractionation and slice electrophysiology recordings confirmed a predominantly presynaptic effect. Together, these data reveal a nonopioid pathway engaged by a human-tested drug to produce antinociception.
Collapse
Affiliation(s)
- Laurent Martin
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
- Department of Anesthesiology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Mohab Ibrahim
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
- Neuroscience Graduate Interdisciplinary Program, College of Medicine, University of Arizona, Tucson, AZ 85742, USA
| | - Kimberly Gomez
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Jie Yu
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Song Cai
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Lindsey A. Chew
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Shreya Sai Bellampalli
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Aubin Moutal
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Tally Largent-Milnes
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
- Neuroscience Graduate Interdisciplinary Program, College of Medicine, University of Arizona, Tucson, AZ 85742, USA
| | - Frank Porreca
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
- Department of Anesthesiology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
- Neuroscience Graduate Interdisciplinary Program, College of Medicine, University of Arizona, Tucson, AZ 85742, USA
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, Arizona 85724, USA
| | | | - Amol Patwardhan
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
- Department of Anesthesiology, College of Medicine, University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
12
|
Fukasawa T, Yoshizaki-Ogawa A, Enomoto A, Miyagawa K, Sato S, Yoshizaki A. Pharmacotherapy of Itch-Antihistamines and Histamine Receptors as G Protein-Coupled Receptors. Int J Mol Sci 2022; 23:6579. [PMID: 35743023 PMCID: PMC9223628 DOI: 10.3390/ijms23126579] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/05/2022] [Accepted: 06/07/2022] [Indexed: 02/05/2023] Open
Abstract
Itching can decrease quality of life and exacerbate skin symptoms due to scratching. Itching not only contributes to disease progression but also triggers complications such as skin infections and eye symptoms. Therefore, controlling itching is very important in therapeutic management. In addition to the well-known histamine, IL-31, IL-4 and IL-13 have recently been reported as factors that induce itching. Itching may also be caused by factors other than these histamines. However, we do not know the extent to which these factors are involved in each disease. In addition, the degree of involvement is likely to vary among individuals. To date, antihistamines have been widely used to treat itching and are often effective, suggesting that histamine is more or less involved in itchy diseases. This review discusses the ligand-receptor perspective and describes the dynamics of G protein-coupled receptors, their role as biased agonists, their role as inverse agonists, proactive antihistamine therapy, and drug selection with consideration of impaired performance and anti-PAF effects.
Collapse
Affiliation(s)
- Takemichi Fukasawa
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; (T.F.); (A.Y.-O.); (S.S.)
| | - Asako Yoshizaki-Ogawa
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; (T.F.); (A.Y.-O.); (S.S.)
| | - Atsushi Enomoto
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; (A.E.); (K.M.)
| | - Kiyoshi Miyagawa
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; (A.E.); (K.M.)
| | - Shinichi Sato
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; (T.F.); (A.Y.-O.); (S.S.)
| | - Ayumi Yoshizaki
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan; (T.F.); (A.Y.-O.); (S.S.)
| |
Collapse
|
13
|
Paul S, Dinesh Kumar SM, Syamala SS, Balakrishnan S, Vijayan V, Arumugaswami V, Sudhakar S. Identification, tissue specific expression analysis and functional characterization of arrestin gene (ARRDC) in the earthworm Eudrilus eugeniae: a molecular hypothesis behind worm photoreception. Mol Biol Rep 2022; 49:4225-4236. [PMID: 35211863 DOI: 10.1007/s11033-022-07256-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/09/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND The arrestin domain containing proteins (ARRDCs) are crucial adaptor proteins assist in signal transduction and regulation of sensory physiology. The molecular localization of the ARRDC gene has been confined mainly to the mammalian system while in invertebrates the expression pattern was not addressed significantly. The present study reports the identification, tissue specific expression and functional characterization of an ARRDC transcript in earthworm, Eudrilus eugeniae. METHODS AND RESULTS The coding region of earthworm ARRDC transcript was 1146 bp in length and encoded a protein of 381 amino acid residues. The worm ARRDC protein consists of conserved N-terminal and C-terminal regions and showed significant homology with the ARRDC3 sequence of other species. The tissue specific expression analysis through whole mount in-situ hybridization denoted the expression of ARRDC transcript in the central nervous system of the worm which includes cerebral ganglion and ventral nerve cord. Besides, the expression of ARRDC gene was observed in the epidermal region of earthworm skin. The functional characterization of ARRDC gene was assessed through siRNA silencing and the gene was found to play key role in the light sensing ability and photophobic movement of the worm. CONCLUSIONS The neuronal and dermal expression patterns of ARRDC gene and its functional characterization hypothesized the role of the gene in assisting the photosensory cells to regulate the process of photoreception and phototransduction in the worm.
Collapse
Affiliation(s)
- Sayan Paul
- Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu, 627012, India
- Centre for Cardiovascular Biology and Disease, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, 560065, India
| | - Sudalai Mani Dinesh Kumar
- Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu, 627012, India
| | - Sandhya Soman Syamala
- Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu, 627012, India
| | | | - Vijithkumar Vijayan
- Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu, 627012, India
| | | | - Sivasubramaniam Sudhakar
- Department of Biotechnology, Manonmaniam Sundaranar University, Tirunelveli, Tamil Nadu, 627012, India.
| |
Collapse
|
14
|
Drube J, Haider RS, Matthees ESF, Reichel M, Zeiner J, Fritzwanker S, Ziegler C, Barz S, Klement L, Filor J, Weitzel V, Kliewer A, Miess-Tanneberg E, Kostenis E, Schulz S, Hoffmann C. GPCR kinase knockout cells reveal the impact of individual GRKs on arrestin binding and GPCR regulation. Nat Commun 2022; 13:540. [PMID: 35087057 PMCID: PMC8795447 DOI: 10.1038/s41467-022-28152-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 01/06/2022] [Indexed: 12/19/2022] Open
Abstract
G protein-coupled receptors (GPCRs) activate G proteins and undergo a complex regulation by interaction with GPCR kinases (GRKs) and the formation of receptor-arrestin complexes. However, the impact of individual GRKs on arrestin binding is not clear. We report the creation of eleven combinatorial HEK293 knockout cell clones lacking GRK2/3/5/6, including single, double, triple and the quadruple GRK knockout. Analysis of β-arrestin1/2 interactions for twelve GPCRs in our GRK knockout cells enables the differentiation of two main receptor subsets: GRK2/3-regulated and GRK2/3/5/6-regulated receptors. Furthermore, we identify GPCRs that interact with β-arrestins via the overexpression of specific GRKs even in the absence of agonists. Finally, using GRK knockout cells, PKC inhibitors and β-arrestin mutants, we present evidence for differential receptor-β-arrestin1/2 complex configurations mediated by selective engagement of kinases. We anticipate our GRK knockout platform to facilitate the elucidation of previously unappreciated details of GRK-specific GPCR regulation and β-arrestin complex formation.
Collapse
Affiliation(s)
- J Drube
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745, Jena, Germany
| | - R S Haider
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745, Jena, Germany
| | - E S F Matthees
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745, Jena, Germany
| | - M Reichel
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745, Jena, Germany
| | - J Zeiner
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | - S Fritzwanker
- Institut für Pharmakologie und Toxikologie, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Drackendorfer Straße 1, D-07747, Jena, Germany
| | - C Ziegler
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745, Jena, Germany
| | - S Barz
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745, Jena, Germany
| | - L Klement
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745, Jena, Germany
| | - J Filor
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745, Jena, Germany
| | - V Weitzel
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745, Jena, Germany
| | - A Kliewer
- Institut für Pharmakologie und Toxikologie, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Drackendorfer Straße 1, D-07747, Jena, Germany
| | - E Miess-Tanneberg
- Institut für Pharmakologie und Toxikologie, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Drackendorfer Straße 1, D-07747, Jena, Germany
| | - E Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Nussallee 6, 53115, Bonn, Germany
| | - S Schulz
- Institut für Pharmakologie und Toxikologie, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Drackendorfer Straße 1, D-07747, Jena, Germany
| | - C Hoffmann
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745, Jena, Germany.
| |
Collapse
|
15
|
Suo WZ. GRK5 Deficiency Causes Mild Cognitive Impairment due to Alzheimer's Disease. J Alzheimers Dis 2021; 85:1399-1410. [PMID: 34958040 DOI: 10.3233/jad-215379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Prevention of Alzheimer's disease (AD) is a high priority mission while searching for a disease modifying therapy for AD, a devastating major public health crisis. Clinical observations have identified a prodromal stage of AD for which the patients have mild cognitive impairment (MCI) though do not yet meet AD diagnostic criteria. As an identifiable transitional stage before the onset of AD, MCI should become the high priority target for AD prevention, assuming successful prevention of MCI and/or its conversion to AD also prevents the subsequent AD. By pulling this string, one demonstrated cause of amnestic MCI appears to be the deficiency of G protein-coupled receptor-5 (GRK5). The most compelling evidence is that GRK5 knockout (GRK5KO) mice naturally develop into aMCI during aging. Moreover, GRK5 deficiency was reported to occur during prodromal stage of AD in CRND8 transgenic mice. When a GRK5KO mouse was crossbred with Tg2576 Swedish amyloid precursor protein transgenic mouse, the resulted double transgenic GAP mice displayed exaggerated behavioral and pathological changes across the spectrum of AD pathogenesis. Therefore, the GRK5 deficiency possesses unique features and advantage to serve as a prophylactic therapeutic target for MCI due to AD.
Collapse
Affiliation(s)
- William Z Suo
- Laboratory for Alzheimer's Disease & Aging Research, VA Medical Center, Kansas City, MO, USA.,Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA.,The University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA
| |
Collapse
|
16
|
Wang A, Chen W, Tao S. Genome-wide characterization, evolution, structure, and expression analysis of the F-box genes in Caenorhabditis. BMC Genomics 2021; 22:889. [PMID: 34895149 PMCID: PMC8665587 DOI: 10.1186/s12864-021-08189-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 11/19/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND F-box proteins represent a diverse class of adaptor proteins of the ubiquitin-proteasome system (UPS) that play critical roles in the cell cycle, signal transduction, and immune response by removing or modifying cellular regulators. Among closely related organisms of the Caenorhabditis genus, remarkable divergence in F-box gene copy numbers was caused by sizeable species-specific expansion and contraction. Although F-box gene number expansion plays a vital role in shaping genomic diversity, little is known about molecular evolutionary mechanisms responsible for substantial differences in gene number of F-box genes and their functional diversification in Caenorhabditis. Here, we performed a comprehensive evolution and underlying mechanism analysis of F-box genes in five species of Caenorhabditis genus, including C. brenneri, C. briggsae, C. elegans, C. japonica, and C. remanei. RESULTS Herein, we identified and characterized 594, 192, 377, 39, 1426 F-box homologs encoding putative F-box proteins in the genome of C. brenneri, C. briggsae, C. elegans, C. japonica, and C. remanei, respectively. Our work suggested that extensive species-specific tandem duplication followed by a small amount of gene loss was the primary mechanism responsible for F-box gene number divergence in Caenorhabditis genus. After F-box gene duplication events occurred, multiple mechanisms have contributed to gene structure divergence, including exon/intron gain/loss, exonization/pseudoexonization, exon/intron boundaries alteration, exon splits, and intron elongation by tandem repeats. Based on high-throughput RNA sequencing data analysis, we proposed that F-box gene functions have diversified by sub-functionalization through highly divergent stage-specific expression patterns in Caenorhabditis species. CONCLUSIONS Massive species-specific tandem duplications and occasional gene loss drove the rapid evolution of the F-box gene family in Caenorhabditis, leading to complex gene structural variation and diversified functions affecting growth and development within and among Caenorhabditis species. In summary, our findings outline the evolution of F-box genes in the Caenorhabditis genome and lay the foundation for future functional studies.
Collapse
Affiliation(s)
- Ailan Wang
- State Key Laboratory of Crop Stress Biology in Arid Areas and College of Life Sciences, Northwest A & F University, Yangling, 712100 Shaanxi China
- Bioinformatics Center, Northwest A&F University, Yangling, Shaanxi China
- Geneis (Beijing) Co., Beijing, China
| | - Wei Chen
- State Key Laboratory of Crop Stress Biology in Arid Areas and College of Life Sciences, Northwest A & F University, Yangling, 712100 Shaanxi China
- Bioinformatics Center, Northwest A&F University, Yangling, Shaanxi China
| | - Shiheng Tao
- State Key Laboratory of Crop Stress Biology in Arid Areas and College of Life Sciences, Northwest A & F University, Yangling, 712100 Shaanxi China
- Bioinformatics Center, Northwest A&F University, Yangling, Shaanxi China
| |
Collapse
|
17
|
Islam MA, Rallabandi VPS, Mohammed S, Srinivasan S, Natarajan S, Dudekula DB, Park J. Screening of β1- and β2-Adrenergic Receptor Modulators through Advanced Pharmacoinformatics and Machine Learning Approaches. Int J Mol Sci 2021; 22:11191. [PMID: 34681845 PMCID: PMC8538848 DOI: 10.3390/ijms222011191] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/14/2021] [Accepted: 10/14/2021] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular diseases (CDs) are a major concern in the human race and one of the leading causes of death worldwide. β-Adrenergic receptors (β1-AR and β2-AR) play a crucial role in the overall regulation of cardiac function. In the present study, structure-based virtual screening, machine learning (ML), and a ligand-based similarity search were conducted for the PubChem database against both β1- and β2-AR. Initially, all docked molecules were screened using the threshold binding energy value. Molecules with a better binding affinity were further used for segregation as active and inactive through ML. The pharmacokinetic assessment was carried out on molecules retained in the above step. Further, similarity searching of the ChEMBL and DrugBank databases was performed. From detailed analysis of the above data, four compounds for each of β1- and β2-AR were found to be promising in nature. A number of critical ligand-binding amino acids formed potential hydrogen bonds and hydrophobic interactions. Finally, a molecular dynamics (MD) simulation study of each molecule bound with the respective target was performed. A number of parameters obtained from the MD simulation trajectories were calculated and substantiated the stability between the protein-ligand complex. Hence, it can be postulated that the final molecules might be crucial for CDs subjected to experimental validation.
Collapse
Affiliation(s)
- Md Ataul Islam
- 3BIGS Omicscore Pvt. Ltd., 1, O Shaughnessy Rd, Langford Gardens, Bengaluru, Karnataka 560025, India; (M.A.I.); (V.P.S.R.); (S.M.); (S.S.); (D.B.D.)
| | - V. P. Subramanyam Rallabandi
- 3BIGS Omicscore Pvt. Ltd., 1, O Shaughnessy Rd, Langford Gardens, Bengaluru, Karnataka 560025, India; (M.A.I.); (V.P.S.R.); (S.M.); (S.S.); (D.B.D.)
| | - Sameer Mohammed
- 3BIGS Omicscore Pvt. Ltd., 1, O Shaughnessy Rd, Langford Gardens, Bengaluru, Karnataka 560025, India; (M.A.I.); (V.P.S.R.); (S.M.); (S.S.); (D.B.D.)
| | - Sridhar Srinivasan
- 3BIGS Omicscore Pvt. Ltd., 1, O Shaughnessy Rd, Langford Gardens, Bengaluru, Karnataka 560025, India; (M.A.I.); (V.P.S.R.); (S.M.); (S.S.); (D.B.D.)
| | | | - Dawood Babu Dudekula
- 3BIGS Omicscore Pvt. Ltd., 1, O Shaughnessy Rd, Langford Gardens, Bengaluru, Karnataka 560025, India; (M.A.I.); (V.P.S.R.); (S.M.); (S.S.); (D.B.D.)
| | - Junhyung Park
- 3BIGS Co., Ltd., 156, Gwanggyo-ro, Yeongtong-gu, Suwon-si 16506, Korea;
| |
Collapse
|
18
|
Umbarkar P, Ejantkar S, Tousif S, Lal H. Mechanisms of Fibroblast Activation and Myocardial Fibrosis: Lessons Learned from FB-Specific Conditional Mouse Models. Cells 2021; 10:cells10092412. [PMID: 34572061 PMCID: PMC8471002 DOI: 10.3390/cells10092412] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 01/26/2023] Open
Abstract
Heart failure (HF) is a leading cause of morbidity and mortality across the world. Cardiac fibrosis is associated with HF progression. Fibrosis is characterized by the excessive accumulation of extracellular matrix components. This is a physiological response to tissue injury. However, uncontrolled fibrosis leads to adverse cardiac remodeling and contributes significantly to cardiac dysfunction. Fibroblasts (FBs) are the primary drivers of myocardial fibrosis. However, until recently, FBs were thought to play a secondary role in cardiac pathophysiology. This review article will present the evolving story of fibroblast biology and fibrosis in cardiac diseases, emphasizing their recent shift from a supporting to a leading role in our understanding of the pathogenesis of cardiac diseases. Indeed, this story only became possible because of the emergence of FB-specific mouse models. This study includes an update on the advancements in the generation of FB-specific mouse models. Regarding the underlying mechanisms of myocardial fibrosis, we will focus on the pathways that have been validated using FB-specific, in vivo mouse models. These pathways include the TGF-β/SMAD3, p38 MAPK, Wnt/β-Catenin, G-protein-coupled receptor kinase (GRK), and Hippo signaling. A better understanding of the mechanisms underlying fibroblast activation and fibrosis may provide a novel therapeutic target for the management of adverse fibrotic remodeling in the diseased heart.
Collapse
Affiliation(s)
- Prachi Umbarkar
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Correspondence: (P.U.); (H.L.); Tel.: +1-205-996-4248 (P.U.); +1-205-996-4219 (H.L.); Fax: +1-205-975-5104 (H.L.)
| | - Suma Ejantkar
- School of Health Professions, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Sultan Tousif
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Hind Lal
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Correspondence: (P.U.); (H.L.); Tel.: +1-205-996-4248 (P.U.); +1-205-996-4219 (H.L.); Fax: +1-205-975-5104 (H.L.)
| |
Collapse
|
19
|
Kuai J, Han C, Wei W. Potential Regulatory Roles of GRK2 in Endothelial Cell Activity and Pathological Angiogenesis. Front Immunol 2021; 12:698424. [PMID: 34335610 PMCID: PMC8320431 DOI: 10.3389/fimmu.2021.698424] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/21/2021] [Indexed: 12/16/2022] Open
Abstract
G protein-coupled receptor (GPCR) kinase 2 (GRK2) is an integrative node in many signaling network cascades. Emerging evidence indicates that GRK2 can interact with a large number of GPCRs and non-GPCR substrates in both kinase-dependent and -independent modes. Some of these pathways are associated with endothelial cell (EC) activity. The active state of ECs is a pivotal factor in angiogenesis. The occurrence and development of some inflammation-related diseases are accompanied by pathological angiogenesis, but there remains a lack of effective targeted treatments. Alterations in the expression and/or localization of GRK2 have been identified in several types of diseases and have been demonstrated to regulate the angiogenesis process in these diseases. GRK2 as a target may be a promising candidate for anti-angiogenesis therapy.
Collapse
Affiliation(s)
| | | | - Wei Wei
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Anhui Medical University, Hefei, China
| |
Collapse
|
20
|
G protein-coupled receptor kinase 5 deletion suppresses synovial inflammation in a murine model of collagen antibody-induced arthritis. Sci Rep 2021; 11:10481. [PMID: 34006987 PMCID: PMC8131379 DOI: 10.1038/s41598-021-90020-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 04/22/2021] [Indexed: 12/28/2022] Open
Abstract
G protein-coupled receptor kinase 5 (GRK5) regulates inflammatory responses via the nuclear factor-kappa B (NF-κB) pathway. This study investigated the functional involvement of GRK5 in the pathogenesis of inflammatory arthritis. Immunohistochemically, rheumatoid arthritis (RA) synovium had a significantly higher proportion of GRK5-positive cells in the synovial lining layer than healthy control synovium. Gene expression and NF-κB activation in lipopolysaccharide-stimulated human SW982 synovial cells were significantly suppressed by silencing of the GRK5 gene. Similarly, GRK5 kinase activity inhibition in human primary RA synovial cells attenuated gene expressions of inflammatory factors. In a murine model of collagen antibody-induced arthritis, arthritis scores and serum IL6 production of GRK5 knockout (GRK5-/-) mice were significantly lower than those of wild-type mice. Histologically, the degree of synovitis and cartilage degeneration in GRK5-/- mice was significantly lower than in wild-type mice. In in vitro analyses using activated murine macrophages and fibroblast-like synoviocytes, gene expression of inflammatory factors and p65 nuclear translocation were significantly lower in GRK5-/- mice compared to wild-type mice. In conclusion, our results suggested that GRK5 is deeply involved in the pathogenesis of inflammatory arthritis, therefore, GRK5 inhibition could be a potential therapeutic target for types of inflammatory arthritis such as RA.
Collapse
|
21
|
Coleman RC, Eguchi A, Lieu M, Roy R, Barr EW, Ibetti J, Lucchese AM, Peluzzo AM, Gresham K, Chuprun JK, Koch WJ. A peptide of the N terminus of GRK5 attenuates pressure-overload hypertrophy and heart failure. Sci Signal 2021; 14:14/676/eabb5968. [PMID: 33785612 DOI: 10.1126/scisignal.abb5968] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Aberrant changes in gene expression underlie the pathogenesis and progression of pressure-overload heart failure, leading to maladaptive cardiac hypertrophy, ventricular remodeling, and contractile dysfunction. Signaling through the G protein Gq triggers maladaptation and heart failure, in part through the activation of G protein-coupled receptor kinase 5 (GRK5). Hypertrophic stimuli induce the accumulation of GRK5 in the nuclei of cardiomyocytes, where it regulates pathological gene expression through multiple transcription factors including NFAT. The nuclear targeting of GRK5 is mediated by an amino-terminal (NT) domain that binds to calmodulin (CaM). Here, we sought to prevent GRK5-mediated pathology in pressure-overload maladaptation and heart failure by expressing in cardiomyocytes a peptide encoding the GRK5 NT (GRK5nt) that encompasses the CaM binding domain. In cultured cardiomyocytes, GRK5nt expression abrogated Gq-coupled receptor-mediated hypertrophy, including attenuation of pathological gene expression and the transcriptional activity of NFAT and NF-κB. We confirmed that GRK5nt bound to and blocked Ca2+-CaM from associating with endogenous GRK5, thereby preventing GRK5 nuclear accumulation after pressure overload. We generated mice that expressed GRKnt in a cardiac-specific fashion (TgGRK5nt mice), which exhibited reduced cardiac hypertrophy, ventricular dysfunction, pulmonary congestion, and cardiac fibrosis after chronic transverse aortic constriction. Together, our data support a role for GRK5nt as an inhibitor of pathological GRK5 signaling that prevents heart failure.
Collapse
Affiliation(s)
- Ryan C Coleman
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Akito Eguchi
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Melissa Lieu
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Rajika Roy
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Eric W Barr
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Jessica Ibetti
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Anna-Maria Lucchese
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Amanda M Peluzzo
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Kenneth Gresham
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - J Kurt Chuprun
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Walter J Koch
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
22
|
Chaudhary PK, Kim S. The GRKs Reactome: Role in Cell Biology and Pathology. Int J Mol Sci 2021; 22:ijms22073375. [PMID: 33806057 PMCID: PMC8036551 DOI: 10.3390/ijms22073375] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 02/06/2023] Open
Abstract
G protein-coupled receptor kinases (GRKs) are protein kinases that function in concert with arrestins in the regulation of a diverse class of G protein-coupled receptors (GPCRs) signaling. Although GRKs and arrestins are key participants in the regulation of GPCR cascades, the complex regulatory mechanisms of GRK expression, its alternation, and their function are not thoroughly understood. Several studies together with the work from our lab in recent years have revealed the critical role of these kinases in various physiological and pathophysiological processes, including cardiovascular biology, inflammation and immunity, neurodegeneration, thrombosis, and hemostasis. A comprehensive understanding of the mechanisms underlying functional interactions with multiple receptor proteins and how these interactions take part in the development of various pathobiological processes may give rise to novel diagnostic and therapeutic strategies. In this review, we summarize the current research linking the role of GRKs to various aspects of cell biology, pathology, and therapeutics, with a particular focus on thrombosis and hemostasis.
Collapse
|
23
|
Benovic JL. Historical Perspective of the G Protein-Coupled Receptor Kinase Family. Cells 2021; 10:555. [PMID: 33806476 PMCID: PMC7999923 DOI: 10.3390/cells10030555] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 01/14/2023] Open
Abstract
Agonist activation of G protein-coupled receptors promotes sequential interaction of the receptor with heterotrimeric G proteins, G protein-coupled receptor kinases (GRKs), and arrestins. GRKs play a central role in mediating the switch from G protein to arrestin interaction and thereby control processes such as receptor desensitization and trafficking and arrestin-mediated signaling. In this review, I provide a historical perspective on some of the early studies that identified the family of GRKs with a primary focus on the non-visual GRKs. These studies included identification, purification, and cloning of the β-adrenergic receptor kinase in the mid- to late-1980s and subsequent cloning and characterization of additional members of the GRK family. This helped to lay the groundwork for ensuing work focused on understanding the structure and function of these important enzymes.
Collapse
Affiliation(s)
- Jeffrey L Benovic
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
24
|
Targeting GRK5 for Treating Chronic Degenerative Diseases. Int J Mol Sci 2021; 22:ijms22041920. [PMID: 33671974 PMCID: PMC7919044 DOI: 10.3390/ijms22041920] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 12/27/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell-surface receptors and they are responsible for the transduction of extracellular signals, regulating almost all aspects of mammalian physiology. These receptors are specifically regulated by a family of serine/threonine kinases, called GPCR kinases (GRKs). Given the biological role of GPCRs, it is not surprising that GRKs are also involved in several pathophysiological processes. Particular importance is emerging for GRK5, which is a multifunctional protein, expressed in different cell types, and it has been found located in single or multiple subcellular compartments. For instance, when anchored to the plasma membrane, GRK5 exerts its canonical function, regulating GPCRs. However, under certain conditions (e.g., pro-hypertrophic stimuli), GRK5 translocates to the nucleus of cells where it can interact with non-GPCR-related proteins as well as DNA itself to promote “non-canonical” signaling, including gene transcription. Importantly, due to these actions, several studies have demonstrated that GRK5 has a pivotal role in the pathogenesis of chronic-degenerative disorders. This is true in the cardiac cells, tumor cells, and neurons. For this reason, in this review article, we will inform the readers of the most recent evidence that supports the importance of targeting GRK5 to prevent the development or progression of cancer, cardiovascular, and neurological diseases.
Collapse
|
25
|
Bledzka KM, Manaserh IH, Grondolsky J, Pfleger J, Roy R, Gao E, Chuprun JK, Koch WJ, Schumacher SM. A peptide of the amino-terminus of GRK2 induces hypertrophy and yet elicits cardioprotection after pressure overload. J Mol Cell Cardiol 2021; 154:137-153. [PMID: 33548241 DOI: 10.1016/j.yjmcc.2021.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 12/14/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022]
Abstract
G protein-coupled receptor (GPCR) kinase 2 (GRK2) expression and activity are elevated early on in response to several forms of cardiovascular stress and are a hallmark of heart failure. Interestingly, though, in addition to its well-characterized role in regulating GPCRs, mounting evidence suggests a GRK2 "interactome" that underlies a great diversity in its functional roles. Several such GRK2 interacting partners are important for adaptive and maladaptive myocyte growth; therefore, an understanding of domain-specific interactions with signaling and regulatory molecules could lead to novel targets for heart failure therapy. Herein, we subjected transgenic mice with cardiac restricted expression of a short, amino terminal fragment of GRK2 (βARKnt) to pressure overload and found that unlike their littermate controls or previous GRK2 fragments, they exhibited an increased left ventricular wall thickness and mass prior to cardiac stress that underwent proportional hypertrophic growth to controls after acute pressure overload. Importantly, despite this enlarged heart, βARKnt mice did not undergo the expected transition to heart failure observed in controls. Further, βARKnt expression limited adverse left ventricular remodeling and increased cell survival signaling. Proteomic analysis to identify βARKnt binding partners that may underlie the improved cardiovascular phenotype uncovered a selective functional interaction of both endogenous GRK2 and βARKnt with AKT substrate of 160 kDa (AS160). AS160 has emerged as a key downstream regulator of insulin signaling, integrating physiological and metabolic cues to couple energy demand to membrane recruitment of Glut4. Our preliminary data indicate that in βARKnt mice, cardiomyocyte insulin signaling is improved during stress, with a coordinate increase in spare respiratory activity and ATP production without metabolite switching. Surprisingly, these studies also revealed a significant decrease in gonadal fat weight, equivalent to human abdominal fat, in male βARKnt mice at baseline and following cardiac stress. These data suggest that the enhanced AS160-mediated signaling in the βARKnt mice may ameliorate pathological cardiac remodeling through direct modulation of insulin signaling within cardiomyocytes, and translate these to beneficial effects on systemic metabolism.
Collapse
Affiliation(s)
- Kamila M Bledzka
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Iyad H Manaserh
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jessica Grondolsky
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jessica Pfleger
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Rajika Roy
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Erhe Gao
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| | - J Kurt Chuprun
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Walter J Koch
- Center for Translational Medicine, Temple University, Philadelphia, PA 19140, USA; Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Sarah M Schumacher
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
26
|
Venkatesh SR, Singh V. G protein-coupled receptors: The choreographers of innate immunity in Caenorhabditis elegans. PLoS Pathog 2021; 17:e1009151. [PMID: 33476324 PMCID: PMC7819600 DOI: 10.1371/journal.ppat.1009151] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Siddharth R. Venkatesh
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
| | - Varsha Singh
- Department of Molecular Reproduction, Development and Genetics, Indian Institute of Science, Bangalore, India
- * E-mail:
| |
Collapse
|
27
|
The Metabolic Role of GRK2 in Insulin Resistance and Associated Conditions. Cells 2021; 10:cells10010167. [PMID: 33467677 PMCID: PMC7830135 DOI: 10.3390/cells10010167] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
Insulin resistance (IRES) is a pathophysiological condition characterized by the reduced response to insulin of several tissues, including myocardial and skeletal muscle. IRES is associated with obesity, glucose intolerance, dyslipidemia, and hypertension, evolves toward type 2 diabetes, and increases the risk of developing cardiovascular diseases. Several studies designed to explore the mechanisms involved in IRES allowed the identification of a multitude of potential molecular targets. Among the most promising, G Protein Coupled Receptor Kinase type 2 (GRK2) appears to be a suitable one given its functional implications in many cellular processes. In this review, we will discuss the metabolic role of GRK2 in those conditions that are characterized by insulin resistance (diabetes, hypertension, heart failure), and the potentiality of its inhibition as a therapeutic strategy to revert both insulin resistance and its associated phenotypes.
Collapse
|
28
|
Yue X, Deng F, Chen J, Yin J, Zheng J, Chen Y, Huang Q, Gao X, Liu Z, Luo J, Müller A, Heidecke H, Riemekasten G, Petersen F, Yu X. Autoantibodies against C5aR1, C3aR1, CXCR3, and CXCR4 are decreased in primary Sjogren's syndrome. Mol Immunol 2021; 131:112-120. [PMID: 33446393 DOI: 10.1016/j.molimm.2020.12.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 12/12/2020] [Accepted: 12/20/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND Networks formed of numerous autoantibodies (aabs) directed against G-protein coupled receptors (GPCR) have been suggested to play important role in autoimmune disorders. In present study, we aimed to evaluate the association between anti-GPCR antibodies and primary Sjogren's syndrome (pSS) to determine the potential pathogenic factors. METHODS By applying a cell membrane-based ELISA technique, which is capable of detecting aabs against conformational epitopes within GPCR, serum levels of fourteen GPCR were determined in well-characterized patients with pSS (n = 52) and gender-matched healthy controls (n = 54). Comparisons between groups were analyzed by two-tailed Mann-Whitney U test, Bonferroni correction was applied for multiple comparisons. Spearman`s rank correlation coefficients were calculated between variables and visualized by heat map. RESULTS Compared to healthy subjects, sera of patients with pSS showed significantly higher binding to β2AR and ETAR, but lower binding to C5aR1, C3aR1, CXCR3, and CXCR4. Autoantibodies against C5aR1, C3aR1, CXCR3, and CXCR4 were also decreased in patients with rheumatoid arthritis. In pSS patients, levels of anti-CXCR3 and anti-CXCR4 antibodies were negatively correlated with circulating lymphocyte counts. Furthermore, correlation signatures of anti-GPCR antibodies changed dramatically in the patients with pulmonary involvement. CONCLUSIONS This study demonstrates an association between pSS and autoantibodies recognizing GPCR, especially those functionally involved in immune cell migration and exocrine glandular secretion.
Collapse
Affiliation(s)
- Xiaoyang Yue
- Priority Area Asthma & Allergy, Research Center Borstel, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Fengyuan Deng
- Laboratory of Autoimmunity, Medical College of Xiamen University, Xiamen, 361005, China
| | - Juan Chen
- Rheumatology Department of the First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
| | - Junping Yin
- Priority Area Asthma & Allergy, Research Center Borstel, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Junfeng Zheng
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, XinXiang, China
| | - Yan Chen
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, XinXiang, China
| | - Qiaoniang Huang
- Laboratory of Autoimmunity, Medical College of Xiamen University, Xiamen, 361005, China
| | - Xing Gao
- Department of Clinical Laboratory, Xiamen University Hospital, Xiamen University, Xiamen, China
| | - Zuguo Liu
- Eye Institute and Affiliated Xiamen Eye Center of Xiamen University, Xiamen, Fujian, China
| | - Jiao Luo
- Department of Rheumatology & Clinical Immunology, University Hospital of Schleswig-Holstein, Campus Lübeck & University of Lübeck, Lübeck, Germany
| | - Antje Müller
- Department of Rheumatology & Clinical Immunology, University Hospital of Schleswig-Holstein, Campus Lübeck & University of Lübeck, Lübeck, Germany
| | | | - Gabriela Riemekasten
- Department of Rheumatology & Clinical Immunology, University Hospital of Schleswig-Holstein, Campus Lübeck & University of Lübeck, Lübeck, Germany
| | - Frank Petersen
- Priority Area Asthma & Allergy, Research Center Borstel, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Xinhua Yu
- Priority Area Asthma & Allergy, Research Center Borstel, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany.
| |
Collapse
|
29
|
Pivonello C, Patalano R, Negri M, Pirchio R, Colao A, Pivonello R, Auriemma RS. Resistance to Dopamine Agonists in Pituitary Tumors: Molecular Mechanisms. Front Endocrinol (Lausanne) 2021; 12:791633. [PMID: 35095761 PMCID: PMC8789681 DOI: 10.3389/fendo.2021.791633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/16/2021] [Indexed: 11/24/2022] Open
Abstract
Pituitary neuroendocrine tumors (PitNET) are commonly benign tumors accounting for 10-25% of intracranial tumors. Prolactin-secreting adenomas represent the most predominant type of all PitNET and for this subtype of tumors, the medical therapy relies on the use of dopamine agonists (DAs). DAs yield an excellent therapeutic response in reducing tumor size and hormonal secretion targeting the dopamine receptor type 2 (D2DR) whose higher expression in prolactin-secreting adenomas compared to other PitNET is now well established. Moreover, although DAs therapy does not represent the first-line therapy for other PitNET, off-label use of DAs is considered in PitNET expressing D2DR. Nevertheless, DAs primary or secondary resistance, occurring in a subset of patients, may involve several molecular mechanisms, presently not fully elucidated. Dopamine receptors (DRs) expression is a prerequisite for a proper DA function in PitNET and several molecular events may negatively modify DR membrane expression, through the DRs down-regulation and intracellular trafficking, and DR signal transduction pathway. The current mini-review will summarise the presently known molecular events that underpin the unsuccessful therapy with DAs.
Collapse
Affiliation(s)
- Claudia Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università di Napoli (Federico II), Naples, Italy
- *Correspondence: Claudia Pivonello, ;
| | - Roberta Patalano
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università di Napoli (Federico II), Naples, Italy
- Dipartimento di Sanità Pubblica, Università di Napoli (Federico II), Naples, Italy
| | - Mariarosaria Negri
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università di Napoli (Federico II), Naples, Italy
| | - Rosa Pirchio
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università di Napoli (Federico II), Naples, Italy
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università di Napoli (Federico II), Naples, Italy
- United Nations Educational, Scientific and Cultural Organization (UNESCO) Chair for Health Education and Sustainable Development, Federico II University, Naples, Italy
| | - Rosario Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università di Napoli (Federico II), Naples, Italy
- United Nations Educational, Scientific and Cultural Organization (UNESCO) Chair for Health Education and Sustainable Development, Federico II University, Naples, Italy
| | - Renata Simona Auriemma
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università di Napoli (Federico II), Naples, Italy
| |
Collapse
|
30
|
van Gastel J, Leysen H, Boddaert J, Vangenechten L, Luttrell LM, Martin B, Maudsley S. Aging-related modifications to G protein-coupled receptor signaling diversity. Pharmacol Ther 2020; 223:107793. [PMID: 33316288 DOI: 10.1016/j.pharmthera.2020.107793] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023]
Abstract
Aging is a highly complex molecular process, affecting nearly all tissue systems in humans and is the highest risk factor in developing neurodegenerative disorders such as Alzheimer's and Parkinson's disease, cardiovascular disease and Type 2 diabetes mellitus. The intense complexity of the aging process creates an incentive to develop more specific drugs that attenuate or even reverse some of the features of premature aging. As our current pharmacopeia is dominated by therapeutics that target members of the G protein-coupled receptor (GPCR) superfamily it may be prudent to search for effective anti-aging therapeutics in this fertile domain. Since the first demonstration of GPCR-based β-arrestin signaling, it has become clear that an enhanced appreciation of GPCR signaling diversity may facilitate the creation of therapeutics with selective signaling activities. Such 'biased' ligand signaling profiles can be effectively investigated using both standard molecular biological techniques as well as high-dimensionality data analyses. Through a more nuanced appreciation of the quantitative nature across the multiple dimensions of signaling bias that drugs possess, researchers may be able to further refine the efficacy of GPCR modulators to impact the complex aberrations that constitute the aging process. Identifying novel effector profiles could expand the effective pharmacopeia and assist in the design of precision medicines. This review discusses potential non-G protein effectors, and specifically their potential therapeutic suitability in aging and age-related disorders.
Collapse
Affiliation(s)
- Jaana van Gastel
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium
| | - Hanne Leysen
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium
| | - Jan Boddaert
- Molecular Pathology Group, Faculty of Medicine and Health Sciences, Laboratory of Cell Biology and Histology, Antwerp, Belgium
| | - Laura Vangenechten
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Louis M Luttrell
- Division of Endocrinology, Diabetes & Medical Genetics, Medical University of South Carolina, USA
| | - Bronwen Martin
- Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium
| | - Stuart Maudsley
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Faculty of Pharmacy, Biomedical and Veterinary Science, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
31
|
Yang A, Yu G, Wu Y, Wang H. Role of β2-adrenergic receptors in chronic obstructive pulmonary disease. Life Sci 2020; 265:118864. [PMID: 33301808 DOI: 10.1016/j.lfs.2020.118864] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/19/2020] [Accepted: 11/29/2020] [Indexed: 01/14/2023]
Abstract
Beta-2 adrenergic receptors (β2-ARs) have important roles in the pathogenesis and treatment of chronic obstructive pulmonary disease (COPD). In recent years, progress has been made in the study of β2-ARs. Here, we introduce the basic concepts of β2-ARs, related pathways, as well as application of blockers/agonists of β2-ARs, and β2-AR autoantibodies in COPD. Drugs targeting the β2-AR are being developed rapidly, and we expect them to improve the symptoms and prognosis of COPD patients in the future.
Collapse
Affiliation(s)
- Ailin Yang
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xicheng District, Beijing 100050, China
| | - Ganggang Yu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xicheng District, Beijing 100050, China
| | - Yanjun Wu
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xicheng District, Beijing 100050, China.
| | - Haoyan Wang
- Department of Respiratory Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xicheng District, Beijing 100050, China.
| |
Collapse
|
32
|
Luo QJ, Sun MX, Guo YW, Tan SW, Wu XY, Abassa KK, Lin L, Liu HL, Jiang J, Wei XQ. Sodium butyrate protects against lipopolysaccharide-induced liver injury partially via the GPR43/ β-arrestin-2/NF-κB network. Gastroenterol Rep (Oxf) 2020; 9:154-165. [PMID: 34026223 PMCID: PMC8128024 DOI: 10.1093/gastro/goaa085] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 10/11/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023] Open
Abstract
Background Butyrate acts as a regulator in multiple inflammatory organ injuries. However, the role of butyrate in acute liver injury has not yet been fully explored. In the present study, we aimed to investigate the association between butyrate and lipopolysaccharide (LPS)-induced acute liver injury and the signaling pathways involved. Methods LPS-induced acute liver injury was induced by intraperitoneal injection of LPS (5 mg/kg) in G-protein-coupled receptor 43 (GPR43)-knockout (KO) and wild-type female C57BL/6 mice. Sodium butyrate (500mg/kg) was administered intraperitoneally 30 min prior to LPS exposure. Liver injury was detected by serum markers, tissue morphology, and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL). Pro-inflammatory-factor levels were detected by enzyme-linked immunosorbent assay and real-time polymerase chain reaction (RT-PCR). Cell models were first treated with sodium butyrate (4 μmol/mL), followed by LPS (1 μg/mL) half an hour later in GPR43 small interfering RNA (siRNA)-transfected or control RAW264.7 cells. Cell-inflammation status was evaluated through detecting pro-inflammatory-factor expression by RT-PCR and also through checking toll-like receptor 4/nuclear factor-κB (TLR4/NF-κB)-element levels including TLR4, TRAF6, IKKβ, IкBα, phospho-IкBα, p65, and phospho-p65 by Western blot. The interaction between GPR43 and β-arrestin-2 was tested by co-immunoprecipitation. Results Sodium butyrate reversed the LPS-induced tissue-morphology changes and high levels of serum alanine aminotransferase, aspartate transaminase, myeloperoxidase, TUNEL, and pro-inflammatory cytokines such as tumor necrosis factor-α and interleukin-6. The ameliorating effect of sodium butyrate was weakened in GPR43-KO mice and GPR43 siRNA RAW264.7 cells, compared with those of GPR43-positive controls. Sodium butyrate downregulated some elements of the TLR4/NF-κB pathway, including phospho-IκBα and phospho-p65, in RAW264.7 cells. Increased interactions between GPR43 and β-arrestin-2, and between β-arrestin-2 and IкBα were observed. Conclusion Sodium butyrate significantly attenuated LPS-induced liver injury by reducing the inflammatory response partially via the GPR43/β-arrestin-2/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Qian-Jiang Luo
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China.,Department of Gastroenterology, The Eighth Affiliated Hospital of Sun Yat-sen University (Shenzhen Futian Hospital), Shenzhen, Guangdong, P. R. China
| | - Mei-Xing Sun
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Yun-Wei Guo
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Si-Wei Tan
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Xiao-Ying Wu
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Kodjo-Kunale Abassa
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Li Lin
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Hui-Ling Liu
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Jie Jiang
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Xiu-Qing Wei
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
33
|
Zhang D, Wang Y, Lin H, Sun Y, Wang M, Jia Y, Yu X, Jiang H, Xu W, Sun JP, Xu Z. Function and therapeutic potential of G protein-coupled receptors in epididymis. Br J Pharmacol 2020; 177:5489-5508. [PMID: 32901914 DOI: 10.1111/bph.15252] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 08/08/2020] [Accepted: 09/03/2020] [Indexed: 12/14/2022] Open
Abstract
Infertility rates for both females and males have increased continuously in recent years. Currently, effective treatments for male infertility with defined mechanisms or targets are still lacking. G protein-coupled receptors (GPCRs) are the largest class of drug targets, but their functions and the implications for the therapeutic development for male infertility largely remain elusive. Nevertheless, recent studies have shown that several members of the GPCR superfamily play crucial roles in the maintenance of ion-water homeostasis of the epididymis, development of the efferent ductules, formation of the blood-epididymal barrier and maturation of sperm. Knowledge of the functions, genetic variations and working mechanisms of such GPCRs, along with the drugs and ligands relevant to their specific functions, provide future directions and a great arsenal for new developments in the treatment of male infertility.
Collapse
Affiliation(s)
- Daolai Zhang
- Department of Pharmaceutical Sciences, Binzhou Medical University, Yantai, China.,Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, China
| | - Yanfei Wang
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Hui Lin
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, China
| | - Yujing Sun
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, China
| | - Mingwei Wang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, China
| | - Yingli Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Xiao Yu
- Department of Physiology, School of Medicine, Shandong University, Jinan, China
| | - Hui Jiang
- Department of Urology, Peking University Third Hospital, Beijing, China.,Department of Reproductive Medicine Center, Peking University Third Hospital, Beijing, China
| | - Wenming Xu
- Joint Laboratory of Reproductive Medicine, SCU-CUHK, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University West China Second University Hospital, Chengdu, China
| | - Jin-Peng Sun
- Department of Pharmaceutical Sciences, Binzhou Medical University, Yantai, China.,Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, School of Medicine, Shandong University, Jinan, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking University, Beijing, China
| | - Zhigang Xu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China.,Shandong Provincial Collaborative Innovation Center of Cell Biology, Shandong Normal University, Jinan, China
| |
Collapse
|
34
|
Zhang J, Vardy E, Muise ES, Wang TM, Visconti R, Vadlamudi A, Pinto S, Peier AM. Utilizing Designed Receptors Exclusively Activated by Designer Drug Chemogenetic Tools to Identify Beneficial G Protein-Coupled Receptor Signaling for Fibrosis. J Pharmacol Exp Ther 2020; 375:357-366. [PMID: 32848074 DOI: 10.1124/jpet.120.000103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/04/2020] [Indexed: 12/21/2022] Open
Abstract
Fibrosis or accumulation of extracellular matrix is an evolutionarily conserved mechanism adopted by an organism as a response to chronic injury. Excessive fibrosis, however, leads to disruption of organ homeostasis and is a common feature of many chronic diseases. G protein-coupled receptors (GPCRs) are important cell signaling mediators and represent molecular targets for many Food and Drug Administration-approved drugs. To identify new targets for fibrosis, we used a synthetic GPCR system named designed receptors exclusively activated by designer drugs (DREADDs) to probe signaling pathways essential for fibrotic response. We found that upon expression in human lung fibroblasts, activation of Gq- and Gs-DREADDs abrogated the induction of TGFβ-induced fibrosis marker genes. Genome-wide transcriptome analysis identified dysregulation of multiple GPCRs in lung fibroblasts treated with TGFβ To investigate endogenous GPCR modulating TGFβ signaling, we selected 13 GPCRs that signal through Gq or Gs and activated them by using specific agonists. We examined the impact of each agonist and how activation of endogenous GPCR affects TGFβ signaling. Among the agonists examined, prostaglandin receptor agonists demonstrated the strongest inhibitory effect on fibrosis. Together, we have demonstrated that the DREADDs system is a valuable tool to identify beneficial GPCR signaling for fibrosis. This study in fibroblasts has served as a proof of concept and allowed us to further develop in vivo models for fibrosis GPCR discovery. SIGNIFICANCE STATEMENT: Fibrosis is the hallmark of many end-stage cardiometabolic diseases, and there is an unmet medical need to discover new antifibrotic therapies, reduce disease progression, and bring clinically meaningful efficacy to patients. Our work utilizes designed receptors exclusively activated by designer drug chemogenetic tools to identify beneficial GPCR signaling for fibrosis, providing new insights into GPCR drug discovery.
Collapse
Affiliation(s)
- Ji Zhang
- Departments of Cardiometabolic Diseases (J.Z., S.P.), Screening and Compound Profiling (E.V., R.V., A.V., A.M.P.), GpGx (E.S.M.), and Translational Biomarkers (T.-M.W.), MRL, Merck & Co., Inc., Kenilworth, New Jersey; and Kallyope Inc., New York, New York (E.V., S.P.)
| | - Eyal Vardy
- Departments of Cardiometabolic Diseases (J.Z., S.P.), Screening and Compound Profiling (E.V., R.V., A.V., A.M.P.), GpGx (E.S.M.), and Translational Biomarkers (T.-M.W.), MRL, Merck & Co., Inc., Kenilworth, New Jersey; and Kallyope Inc., New York, New York (E.V., S.P.)
| | - Eric S Muise
- Departments of Cardiometabolic Diseases (J.Z., S.P.), Screening and Compound Profiling (E.V., R.V., A.V., A.M.P.), GpGx (E.S.M.), and Translational Biomarkers (T.-M.W.), MRL, Merck & Co., Inc., Kenilworth, New Jersey; and Kallyope Inc., New York, New York (E.V., S.P.)
| | - Tzu-Ming Wang
- Departments of Cardiometabolic Diseases (J.Z., S.P.), Screening and Compound Profiling (E.V., R.V., A.V., A.M.P.), GpGx (E.S.M.), and Translational Biomarkers (T.-M.W.), MRL, Merck & Co., Inc., Kenilworth, New Jersey; and Kallyope Inc., New York, New York (E.V., S.P.)
| | - Richard Visconti
- Departments of Cardiometabolic Diseases (J.Z., S.P.), Screening and Compound Profiling (E.V., R.V., A.V., A.M.P.), GpGx (E.S.M.), and Translational Biomarkers (T.-M.W.), MRL, Merck & Co., Inc., Kenilworth, New Jersey; and Kallyope Inc., New York, New York (E.V., S.P.)
| | - Ashita Vadlamudi
- Departments of Cardiometabolic Diseases (J.Z., S.P.), Screening and Compound Profiling (E.V., R.V., A.V., A.M.P.), GpGx (E.S.M.), and Translational Biomarkers (T.-M.W.), MRL, Merck & Co., Inc., Kenilworth, New Jersey; and Kallyope Inc., New York, New York (E.V., S.P.)
| | - Shirly Pinto
- Departments of Cardiometabolic Diseases (J.Z., S.P.), Screening and Compound Profiling (E.V., R.V., A.V., A.M.P.), GpGx (E.S.M.), and Translational Biomarkers (T.-M.W.), MRL, Merck & Co., Inc., Kenilworth, New Jersey; and Kallyope Inc., New York, New York (E.V., S.P.)
| | - Andrea M Peier
- Departments of Cardiometabolic Diseases (J.Z., S.P.), Screening and Compound Profiling (E.V., R.V., A.V., A.M.P.), GpGx (E.S.M.), and Translational Biomarkers (T.-M.W.), MRL, Merck & Co., Inc., Kenilworth, New Jersey; and Kallyope Inc., New York, New York (E.V., S.P.)
| |
Collapse
|
35
|
Galkov M, Kiseleva E, Gulyaev M, Sidorova M, Gorbacheva L. New PAR1 Agonist Peptide Demonstrates Protective Action in a Mouse Model of Photothrombosis-Induced Brain Ischemia. Front Neurosci 2020; 14:335. [PMID: 32547356 PMCID: PMC7273131 DOI: 10.3389/fnins.2020.00335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 03/20/2020] [Indexed: 11/23/2022] Open
Abstract
Protease-activated receptors (PARs) are involved not only in hemostasis but also in the development of ischemic brain injury. In the present work, we examined in vivo effects of a new peptide (AP9) composing Asn47-Phen55 of PAR1 “tethered ligand” generated by activated protein C. We chose a mouse model of photothrombosis (PT)-induced ischemia to assess AP9 effects in vivo. To reveal the molecular mechanism of AP9 action, mice lacking β-arrestin-2 were used. AP9 was injected intravenously once 10 min before PT at doses of 0.2, 2, or 20 mg/kg, or twice, that is, 10 min before and 1 h after PT at a dose of 20 mg/kg. Lesion volume was measured by magnetic resonance imaging and staining of brain sections with tetrazolium salt. Neurologic deficit was estimated using the cylinder and the grid-walk tests. Blood–brain barrier (BBB) disruption was assessed by Evans blue dye extraction. Eosin-hematoxylin staining and immunohistochemical staining were applied to evaluate the number of undamaged neurons and activated glial cells in the penumbra. A single administration of AP9 (20 mg/kg), as well as its two injections (20 mg/kg), decreased brain lesion volume. A double administration of AP9 also reduced BBB disruption and neurological deficit in mice. We did not observe the protective effect of AP9 in mice lacking β-arrestin-2 after PT. Thus, we demonstrated for the first time protective properties of a PAR1 agonist peptide, AP9, in vivo. β-Arrestin-2 was required for the protective action of AP9 in PT-induced brain ischemia.
Collapse
Affiliation(s)
- Maksim Galkov
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia.,Electrophysiology Laboratory, Translational Medicine Institute, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Ekaterina Kiseleva
- Electrophysiology Laboratory, Translational Medicine Institute, Pirogov Russian National Research Medical University, Moscow, Russia.,Department of Cell Biology, Koltzov Institute of Developmental Biology of Russian Academy of Sciences, Moscow, Russia
| | - Mikhail Gulyaev
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Maria Sidorova
- Laboratory of Peptide Synthesis, Institute of Experimental Cardiology, National Medical Research Center for Cardiology of Russian Ministry of Health, Moscow, Russia
| | - Liubov Gorbacheva
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia.,Electrophysiology Laboratory, Translational Medicine Institute, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
36
|
Sueishi T, Akasaki Y, Goto N, Kurakazu I, Toya M, Kuwahara M, Uchida T, Hayashida M, Tsushima H, Bekki H, Lotz MK, Nakashima Y. GRK5 Inhibition Attenuates Cartilage Degradation via Decreased NF-κB Signaling. Arthritis Rheumatol 2020; 72:620-631. [PMID: 31696655 DOI: 10.1002/art.41152] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 10/29/2019] [Indexed: 12/26/2022]
Abstract
OBJECTIVE NF-κB-dependent signaling is an important modulator in osteoarthritis (OA), and G protein-coupled receptor kinase 5 (GRK5) regulates the NF-κB pathway. This study was undertaken to investigate the functional involvement of GRK5 in OA pathogenesis. METHODS GRK5 expression in normal and OA human knee joints was analyzed immunohistochemically. Gain- or loss-of-function experiments were performed using human and mouse chondrocytes. OA was induced in GRK5-knockout mice by destabilization of the medial meniscus, and histologic examination was performed. OA was also induced in wild-type mice, which were then treated with an intraarticular injection of amlexanox, a selective GRK5 inhibitor, every 5 days for 8 weeks. RESULTS GRK5 protein expression was increased in human OA cartilage. In vitro, expression levels of OA-related factors and NF-κB transcriptional activation were down-regulated by suppression of the GRK5 gene in human OA chondrocytes (3.49-fold decrease in IL6 [P < 0.01], 2.43-fold decrease in MMP13 [P < 0.01], and 2.66-fold decrease in ADAMTS4 [P < 0.01]). Conversely, GRK5 overexpression significantly increased the expression of OA-related catabolic mediators and NF-κB transcriptional activation. On Western blot analysis, GRK5 deletion reduced IκBα phosphorylation (up to 4.4-fold decrease [P < 0.05]) and decreased p65 nuclear translocation (up to 6.4-fold decrease [P < 0.01]) in mouse chondrocytes. In vivo, both GRK5 deletion and intraarticular amlexanox protected mouse cartilage against OA. CONCLUSION Our results suggest that GRK5 regulates cartilage degradation through a catabolic response mediated by NF-κB signaling, and is a potential target for OA treatment. Furthermore, amlexanox may be a major compound in relevant drugs.
Collapse
Affiliation(s)
- Takuya Sueishi
- Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yukio Akasaki
- Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Norio Goto
- Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Ichiro Kurakazu
- Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Masakazu Toya
- Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Masanari Kuwahara
- Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Taisuke Uchida
- Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | | | | | - Hirofumi Bekki
- Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan, and Scripps Research Institute, San Diego, California
| | | | | |
Collapse
|
37
|
Hattori Y, Michel MC. Editorial: G Protein-Coupled Receptor Kinases (GRKs) and β-Arrestins: New Insights Into Disease Regulators. Front Pharmacol 2020; 10:1654. [PMID: 32047443 PMCID: PMC6997549 DOI: 10.3389/fphar.2019.01654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 12/17/2019] [Indexed: 11/16/2022] Open
Affiliation(s)
- Yuichi Hattori
- The Research Institute of Cancer Prevention, Health Sciences University of Hokkaido, Tobetsu, Japan
| | - Martin C Michel
- Department of Pharmacology, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
38
|
Lee SH, Lee M, Yang H, Cho Y, Hong S, Park TH. Bioelectronic sensor mimicking the human neuroendocrine system for the detection of hypothalamic-pituitary-adrenal axis hormones in human blood. Biosens Bioelectron 2020; 154:112071. [PMID: 32056965 DOI: 10.1016/j.bios.2020.112071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/27/2020] [Accepted: 02/01/2020] [Indexed: 12/15/2022]
Abstract
In the neuroendocrine system, corticotropin-releasing hormone (CRH) and adrenocorticotropic hormone (ACTH) play important roles in the regulation of the hypothalamic-pituitary-adrenal (HPA) system. Disorders of the HPA system lead to physiological problems, such as Addison's disease and Cushing's syndrome. Therefore, detection of CRH and ACTH is essential for diagnosing disorders related to the HPA system. Herein, receptors of the HPA axis were used to construct a bioelectronic sensor system for the detection of CRH and ACTH. The CRH receptor, corticotropin-releasing hormone receptor 1 (CRHR1), and the ACTH receptor, melanocortin 2 receptor (MC2R), were produced using an Escherichia coli expression system, and were reconstituted using nanodisc (ND) technology. The receptor-embedded NDs were immobilized on a floating electrode of a carbon nanotube field-effect transistor (CNT-FET). The constructed sensors sensitively detected CRH and ACTH to a concentration of 1 fM with high selectivity in real time. Furthermore, the reliable detection of CRH and ACTH in human plasma by the developed sensors demonstrated their potential in clinical and practical applications. These results indicate that CRHR1 and MC2R-based bioelectronic sensors can be applied for rapid and efficient detection of CRH and ACTH.
Collapse
Affiliation(s)
- Seung Hwan Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea; Department of Bionano Engineering and Bionanotechnology, Hanyang University, Ansan, 15588, Republic of Korea
| | - Minju Lee
- Department of Physics and Astronomy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Heehong Yang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea; Protein Engineering Laboratory, Discovery Unit, MOGAM Institute for Biomedical Research, Yongin, 16924, Republic of Korea
| | - Youngtak Cho
- Department of Physics and Astronomy, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seunghun Hong
- Department of Physics and Astronomy, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Tai Hyun Park
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
39
|
Effect of mutations on drug resistance of smoothened receptor toward inhibitors probed by molecular modeling. Chem Phys Lett 2020. [DOI: 10.1016/j.cplett.2020.137126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
40
|
Rogers TJ. Bidirectional Regulation of Opioid and Chemokine Function. Front Immunol 2020; 11:94. [PMID: 32076421 PMCID: PMC7006827 DOI: 10.3389/fimmu.2020.00094] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 01/14/2020] [Indexed: 12/15/2022] Open
Abstract
The opioid family of GPCRs consists of the classical opioid receptors, designated μ-, κ-, and δ-opioid receptors, and the orphanin-FQ receptor, and these proteins are expressed on both neuronal and hematopoietic cells. A number of laboratories have reported that an important degree of cross-talk can occur between the opioid receptors and the chemokine and chemokine receptor families. As a part of this, the opioid receptors are known to regulate the expression of certain chemokines and chemokine receptors, including those that possess strong pro-inflammatory activity. At the level of receptor function, it is clear that certain members of the chemokine family can mediate cross-desensitization of the opioid receptors. Conversely, the opioid receptors are all able to induce heterologous desensitization of some of the chemokine receptors. Consequently, activation of one or more of the opioid receptors can selectively cross-desensitize chemokine receptors and regulate chemokine function. These cross-talk processes have significant implications for the inflammatory response, since the regulation of both the recruitment of inflammatory cells, as well as the sensation of pain, can be controlled in this way.
Collapse
Affiliation(s)
- Thomas J Rogers
- Center for Inflammation, Translational and Clinical Lung Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
41
|
Affiliation(s)
- Jing Wang
- Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education of China, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.,Yale Center for ImmunoMetabolism, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Richard A Flavell
- Yale Center for ImmunoMetabolism, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China. .,Department of Immunobiology, Yale University School of Medicine, New Haven, CT, 06520-8055, USA. .,Howard Hughes Medical Institute, Yale University, New Haven, CT, 06520-8055, USA.
| | - Hua-Bing Li
- Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education of China, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China. .,Yale Center for ImmunoMetabolism, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| |
Collapse
|
42
|
Yang X, Li S, Zhao Y, Li S, Zhao T, Tai Y, Zhang B, Wang X, Wang C, Chen J, Wang Q, Zhang L, Xu D, Chang Y, Wei W. GRK2 Mediated Abnormal Transduction of PGE2-EP4-cAMP-CREB Signaling Induces the Imbalance of Macrophages Polarization in Collagen-Induced Arthritis Mice. Cells 2019; 8:cells8121596. [PMID: 31818003 PMCID: PMC6953022 DOI: 10.3390/cells8121596] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/05/2019] [Accepted: 12/06/2019] [Indexed: 12/19/2022] Open
Abstract
Rheumatoid arthritis (RA) is characterized by the massive infiltration of various chronic inflammatory cells in synovia. In synovial fluid of patients with RA, M1 macrophages are dominant among all subtypes of macrophages, the mechanisms of macrophages polarization imbalance in RA has not been fully illuminated. The prostaglandin E2 (PGE2) augments M2 polarization in part via the cyclic adenosine monophosphate (cAMP)-cyclic AMP responsive element binding (CREB) signaling. However, previous study found constant stimulus of PGE2 on fibroblast-like synovial cells of adjuvant arthritis rats induced the decrease of cAMP, which is primarily caused by G protein-coupled receptor kinase 2 (GRK2)-induced EP4 over- desensitization. Whether GRK2 mediated-EP4 over-desensitization reduces the level of cAMP and inhibits M2 polarization in RA is unclear. Here we observed M1 macrophages were dominant in peritoneal macrophages (PMs), bone-marrow-derived macrophages (BMMs) and synovial macrophages of collagen-induced arthritis (CIA) mice. PGE2 stimulated M2 polarization via the EP4-cAMP-CREB in normal mice, while failed to promote M2 polarization in the PMs of CIA mice. Further, we found the EP4 over-desensitization stimulated by PGE2 induced abnormal PGE2-cAMP-CREB signaling as well as the imbalance of macrophage polarization. Targeted disruption of GRK2 in Raw264.7 (RAW) through GRK2 siRNA or CRISPR/Cas9 downregulated the M1 macrophage markers, upregulated the M2 macrophage markers and the EP4 membrane localization. The reduced M1/M2 ratio and increased p-CREB expression were observed in BMMs and PMs of GRK2+/− mice. This study highlighted a novel role of GRK2 in regulating macrophages function in RA and provided new idea for precision treatment of RA.
Collapse
Affiliation(s)
- Xuezhi Yang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China; (X.Y.); (S.L.); (Y.Z.); (S.L.); (T.Z.); (Y.T.); (B.Z.); (X.W.); (C.W.); (J.C.); (Q.W.); (L.Z.)
- Public Health and Preventive Medicine Postdoctoral Research Station of Anhui Medical University, Hefei 230032, China
| | - Susu Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China; (X.Y.); (S.L.); (Y.Z.); (S.L.); (T.Z.); (Y.T.); (B.Z.); (X.W.); (C.W.); (J.C.); (Q.W.); (L.Z.)
| | - Yingjie Zhao
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China; (X.Y.); (S.L.); (Y.Z.); (S.L.); (T.Z.); (Y.T.); (B.Z.); (X.W.); (C.W.); (J.C.); (Q.W.); (L.Z.)
| | - Siyu Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China; (X.Y.); (S.L.); (Y.Z.); (S.L.); (T.Z.); (Y.T.); (B.Z.); (X.W.); (C.W.); (J.C.); (Q.W.); (L.Z.)
| | - Tianjiao Zhao
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China; (X.Y.); (S.L.); (Y.Z.); (S.L.); (T.Z.); (Y.T.); (B.Z.); (X.W.); (C.W.); (J.C.); (Q.W.); (L.Z.)
| | - Yu Tai
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China; (X.Y.); (S.L.); (Y.Z.); (S.L.); (T.Z.); (Y.T.); (B.Z.); (X.W.); (C.W.); (J.C.); (Q.W.); (L.Z.)
| | - Bingjie Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China; (X.Y.); (S.L.); (Y.Z.); (S.L.); (T.Z.); (Y.T.); (B.Z.); (X.W.); (C.W.); (J.C.); (Q.W.); (L.Z.)
| | - Xinwei Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China; (X.Y.); (S.L.); (Y.Z.); (S.L.); (T.Z.); (Y.T.); (B.Z.); (X.W.); (C.W.); (J.C.); (Q.W.); (L.Z.)
| | - Chun Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China; (X.Y.); (S.L.); (Y.Z.); (S.L.); (T.Z.); (Y.T.); (B.Z.); (X.W.); (C.W.); (J.C.); (Q.W.); (L.Z.)
| | - Jingyu Chen
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China; (X.Y.); (S.L.); (Y.Z.); (S.L.); (T.Z.); (Y.T.); (B.Z.); (X.W.); (C.W.); (J.C.); (Q.W.); (L.Z.)
| | - Qingtong Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China; (X.Y.); (S.L.); (Y.Z.); (S.L.); (T.Z.); (Y.T.); (B.Z.); (X.W.); (C.W.); (J.C.); (Q.W.); (L.Z.)
| | - Lingling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China; (X.Y.); (S.L.); (Y.Z.); (S.L.); (T.Z.); (Y.T.); (B.Z.); (X.W.); (C.W.); (J.C.); (Q.W.); (L.Z.)
| | - Dexiang Xu
- Public Health and Preventive Medicine Postdoctoral Research Station of Anhui Medical University, Hefei 230032, China
- Correspondence: (D.X.); (Y.C.); (W.W.); Tel./Fax: +86-551-516-1209 (W.W.)
| | - Yan Chang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China; (X.Y.); (S.L.); (Y.Z.); (S.L.); (T.Z.); (Y.T.); (B.Z.); (X.W.); (C.W.); (J.C.); (Q.W.); (L.Z.)
- Correspondence: (D.X.); (Y.C.); (W.W.); Tel./Fax: +86-551-516-1209 (W.W.)
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine (Anhui Medical University), Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei 230032, China; (X.Y.); (S.L.); (Y.Z.); (S.L.); (T.Z.); (Y.T.); (B.Z.); (X.W.); (C.W.); (J.C.); (Q.W.); (L.Z.)
- Correspondence: (D.X.); (Y.C.); (W.W.); Tel./Fax: +86-551-516-1209 (W.W.)
| |
Collapse
|
43
|
Selheim F, Aasebø E, Ribas C, Aragay AM. An Overview on G Protein-coupled Receptor-induced Signal Transduction in Acute Myeloid Leukemia. Curr Med Chem 2019; 26:5293-5316. [PMID: 31032748 DOI: 10.2174/0929867326666190429153247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 03/22/2019] [Accepted: 04/05/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Acute Myeloid Leukemia (AML) is a genetically heterogeneous disease characterized by uncontrolled proliferation of precursor myeloid-lineage cells in the bone marrow. AML is also characterized by patients with poor long-term survival outcomes due to relapse. Many efforts have been made to understand the biological heterogeneity of AML and the challenges to develop new therapies are therefore enormous. G Protein-coupled Receptors (GPCRs) are a large attractive drug-targeted family of transmembrane proteins, and aberrant GPCR expression and GPCR-mediated signaling have been implicated in leukemogenesis of AML. This review aims to identify the molecular players of GPCR signaling, focusing on the hematopoietic system, which are involved in AML to help developing novel drug targets and therapeutic strategies. METHODS We undertook an exhaustive and structured search of bibliographic databases for research focusing on GPCR, GPCR signaling and expression in AML. RESULTS AND CONCLUSION Many scientific reports were found with compelling evidence for the involvement of aberrant GPCR expression and perturbed GPCR-mediated signaling in the development of AML. The comprehensive analysis of GPCR in AML provides potential clinical biomarkers for prognostication, disease monitoring and therapeutic guidance. It will also help to provide marker panels for monitoring in AML. We conclude that GPCR-mediated signaling is contributing to leukemogenesis of AML, and postulate that mass spectrometrybased protein profiling of primary AML cells will accelerate the discovery of potential GPCR related biomarkers for AML.
Collapse
Affiliation(s)
- Frode Selheim
- The Proteomics Unit at the University of Bergen, Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Elise Aasebø
- The Proteomics Unit at the University of Bergen, Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway.,Department of Clinical Science, University of Bergen, Jonas Lies vei 87, 5021 Bergen, Norway
| | - Catalina Ribas
- Departamento de Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), 28049 Madrid, Spain.,Instituto de Investigación Sanitaria La Princesa, 28006 Madrid, Spain.,CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029 Madrid, Spain
| | - Anna M Aragay
- Departamento de Biologia Celular. Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Spanish National Research Council (CSIC), Baldiri i Reixac, 15, 08028 Barcelona, Spain
| |
Collapse
|
44
|
Penela P, Ribas C, Sánchez-Madrid F, Mayor F. G protein-coupled receptor kinase 2 (GRK2) as a multifunctional signaling hub. Cell Mol Life Sci 2019; 76:4423-4446. [PMID: 31432234 PMCID: PMC6841920 DOI: 10.1007/s00018-019-03274-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/06/2019] [Accepted: 08/12/2019] [Indexed: 12/18/2022]
Abstract
Accumulating evidence indicates that G protein-coupled receptor kinase 2 (GRK2) is a versatile protein that acts as a signaling hub by modulating G protein-coupled receptor (GPCR) signaling and also via phosphorylation or scaffolding interactions with an extensive number of non-GPCR cellular partners. GRK2 multifunctionality arises from its multidomain structure and from complex mechanisms of regulation of its expression levels, activity, and localization within the cell, what allows the precise spatio-temporal shaping of GRK2 targets. A better understanding of the GRK2 interactome and its modulation mechanisms is helping to identify the GRK2-interacting proteins and its substrates involved in the participation of this kinase in different cellular processes and pathophysiological contexts.
Collapse
Affiliation(s)
- Petronila Penela
- Departamento de Biología Molecular, Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, 28049, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029, Madrid, Spain
| | - Catalina Ribas
- Departamento de Biología Molecular, Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, 28049, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029, Madrid, Spain
| | - Francisco Sánchez-Madrid
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029, Madrid, Spain
- Cell-Cell Communication Laboratory, Vascular Pathophysiology Area, Centro Nacional Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain
| | - Federico Mayor
- Departamento de Biología Molecular, Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, 28049, Madrid, Spain.
- Instituto de Investigación Sanitaria La Princesa, 28006, Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares, ISCIII (CIBERCV), 28029, Madrid, Spain.
| |
Collapse
|
45
|
5-HT 2A receptor-dependent phosphorylation of mGlu 2 receptor at Serine 843 promotes mGlu 2 receptor-operated G i/o signaling. Mol Psychiatry 2019; 24:1610-1626. [PMID: 29858599 DOI: 10.1038/s41380-018-0069-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 02/19/2018] [Accepted: 03/13/2018] [Indexed: 12/31/2022]
Abstract
The serotonin 5-HT2A and glutamate mGlu2 receptors continue to attract particular attention, given their implication in psychosis associated with schizophrenia and the mechanism of action of atypical antipsychotics and a new class of antipsychotics, respectively. A large body of evidence indicates a functional crosstalk between both receptors in the brain, but the underlying mechanisms are not entirely elucidated. Here, we have explored the influence of 5-HT2A receptor upon the phosphorylation pattern of mGlu2 receptor in light of the importance of specific phosphorylation events in regulating G protein-coupled receptor signaling and physiological outcomes. Among the five mGlu2 receptor-phosphorylated residues identified in HEK-293 cells, the phosphorylation of Ser843 was enhanced upon mGlu2 receptor stimulation by the orthosteric agonist LY379268 only in cells co-expressing the 5-HT2A receptor. Likewise, administration of LY379268 increased mGlu2 receptor phosphorylation at Ser843 in prefrontal cortex of wild-type mice but not 5-HT2A-/- mice. Exposure of HEK-293 cells co-expressing mGlu2 and 5-HT2A receptors to 5-HT also increased Ser843 phosphorylation state to a magnitude similar to that measured in LY379268-treated cells. In both HEK-293 cells and prefrontal cortex, Ser843 phosphorylation elicited by 5-HT2A receptor stimulation was prevented by the mGlu2 receptor antagonist LY341495, while the LY379268-induced effect was abolished by the 5-HT2A receptor antagonist M100907. Mutation of Ser843 into alanine strongly reduced Gi/o signaling elicited by mGlu2 or 5-HT2A receptor stimulation in cells co-expressing both receptors. Collectively, these findings identify mGlu2 receptor phosphorylation at Ser843 as a key molecular event that underlies the functional crosstalk between both receptors.
Collapse
|
46
|
Zhang M, Gao M, Chen J, Song L, Wei W. CP-25 exerts anti-angiogenic effects on a rat model of adjuvant-induced arthritis by promoting GRK2-induced downregulation of CXCR4-ERK1/2 signaling in endothelial cells. Mol Med Rep 2019; 20:4831-4842. [PMID: 31661133 PMCID: PMC6854590 DOI: 10.3892/mmr.2019.10765] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Accepted: 07/04/2019] [Indexed: 12/29/2022] Open
Abstract
Angiogenesis can produce an invasive and destructive front, also named a pannus, comprised of inflammatory vascular tissue that covers and erodes articular cartilage, subchondral bone and peri‑articular soft tissues in rheumatoid arthritis (RA). Paeoniflorin‑6'‑O‑benzene sulfonate (CP‑25) is a novel ester derivative of paeoniflorin. We previously demonstrated that CP‑25 exerts anti‑inflammatory and immunoregulatory effects. CP‑25 also exhibits a marked therapeutic effect on adjuvant‑induced arthritis (AA), and is able to inhibit synovial and immune cell function, according to our previous study. However, the effect of CP‑25 on angiogenesis remains unclear. In the present study, AA was initiated in Sprague‑Dawley rats via intradermal immunization in the right hind metatarsal footpad with heat‑killed Mycobacterium butyricum in liquid paraffin, and rats were divided into four groups: Normal, AA rat model, CP‑25 (50 mg/kg) and methotrexate (0.5 mg/kg) groups (n=10 rats/group). Subsequently, joint synovium in AA rats was pathologically evaluated by hematoxylin and eosin staining, synovial vascular proliferation was evaluated by immunofluorescence, the synovial expression levels of C‑X‑C motif chemokine ligand 12 (CXCL12) were detected by immunohistochemistry and ELISA, and synovial C‑X‑C chemokine receptor type 4 (CXCR4) was detected by western blotting. The results demonstrated that CP‑25 ameliorated clinical signs and pannus formation in the ankle joint in rats with AA. Furthermore, there was a positive correlation between pannus score and CXCL12 and CXCR4 expression. In addition, the effects of CP‑25 on endothelial cell function and CXCL12/CXCR4 signaling were studied in vitro using human umbilical vein endothelial cells (HUVECs). The results demonstrated that CXCL12 significantly promoted HUVEC proliferation, migration and tube formation, and that CP‑25 could reverse these abnormalities by inhibiting plasma membrane localization of G protein‑coupled receptor kinase 2 (GRK2) in HUVECs. These findings suggested that CP‑25 may markedly inhibit pannus formation in AA. This effect may be associated with a reduction in the plasma membrane localization of GRK2 in endothelial cells, an enhancement of the inhibitory effect of GRK2 on ERK1/2 in the cytoplasm, a reduction in the phosphorylation of ERK1/2 and in the function of HUVECs.
Collapse
Affiliation(s)
- Min Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui 230032, P.R. China
| | - Mei Gao
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui 230032, P.R. China
| | - Jinyu Chen
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui 230032, P.R. China
| | - Lihua Song
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui 230032, P.R. China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
47
|
Abbas S, Marino V, Dell’Orco D, Koch KW. Molecular Recognition of Rhodopsin Kinase GRK1 and Recoverin Is Tuned by Switching Intra- and Intermolecular Electrostatic Interactions. Biochemistry 2019; 58:4374-4385. [DOI: 10.1021/acs.biochem.9b00846] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Seher Abbas
- Department of Neuroscience, Division of Biochemistry, University of Oldenburg, 26111 Oldenburg, Germany
| | - Valerio Marino
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Daniele Dell’Orco
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134 Verona, Italy
| | - Karl-Wilhelm Koch
- Department of Neuroscience, Division of Biochemistry, University of Oldenburg, 26111 Oldenburg, Germany
| |
Collapse
|
48
|
Crudden C, Song D, Cismas S, Trocmé E, Pasca S, Calin GA, Girnita A, Girnita L. Below the Surface: IGF-1R Therapeutic Targeting and Its Endocytic Journey. Cells 2019; 8:cells8101223. [PMID: 31600876 PMCID: PMC6829878 DOI: 10.3390/cells8101223] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 09/30/2019] [Accepted: 10/03/2019] [Indexed: 12/15/2022] Open
Abstract
Ligand-activated plasma membrane receptors follow pathways of endocytosis through the endosomal sorting apparatus. Receptors cluster in clathrin-coated pits that bud inwards and enter the cell as clathrin-coated vesicles. These vesicles travel through the acidic endosome whereby receptors and ligands are sorted to be either recycled or degraded. The traditional paradigm postulated that the endocytosis role lay in signal termination through the removal of the receptor from the cell surface. It is now becoming clear that the internalization process governs more than receptor signal cessation and instead reigns over the entire spatial and temporal wiring of receptor signaling. Governing the localization, the post-translational modifications, and the scaffolding of receptors and downstream signal components established the endosomal platform as the master regulator of receptor function. Confinement of components within or between distinct organelles means that the endosome instructs the cell on how to interpret and translate the signal emanating from any given receptor complex into biological effects. This review explores this emerging paradigm with respect to the cancer-relevant insulin-like growth factor type 1 receptor (IGF-1R) and discusses how this perspective could inform future targeting strategies.
Collapse
Affiliation(s)
- Caitrin Crudden
- Department of Oncology-Pathology, Cellular and Molecular Tumor Pathology, Karolinska Institute, and Karolinska University Hospital, 17164 Stockholm, Sweden.
- Department of Pathology, Cancer Centre Amsterdam, Amsterdam UMC, VU University Medical Centre, 1081 HZ Amsterdam, The Netherlands.
| | - Dawei Song
- Department of Oncology-Pathology, Cellular and Molecular Tumor Pathology, Karolinska Institute, and Karolinska University Hospital, 17164 Stockholm, Sweden.
| | - Sonia Cismas
- Department of Oncology-Pathology, Cellular and Molecular Tumor Pathology, Karolinska Institute, and Karolinska University Hospital, 17164 Stockholm, Sweden.
| | - Eric Trocmé
- Department of Oncology-Pathology, Cellular and Molecular Tumor Pathology, Karolinska Institute, and Karolinska University Hospital, 17164 Stockholm, Sweden.
- St. Erik Eye Hospital, 11282 Stockholm, Sweden.
| | - Sylvya Pasca
- Department of Oncology-Pathology, Cellular and Molecular Tumor Pathology, Karolinska Institute, and Karolinska University Hospital, 17164 Stockholm, Sweden.
| | - George A Calin
- Department of Experimental Therapeutics, The University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA.
- Center for RNA Interference and Non-Coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Ada Girnita
- Department of Oncology-Pathology, Cellular and Molecular Tumor Pathology, Karolinska Institute, and Karolinska University Hospital, 17164 Stockholm, Sweden.
- Dermatology Department, Karolinska University Hospital, 17176 Stockholm, Sweden.
| | - Leonard Girnita
- Department of Oncology-Pathology, Cellular and Molecular Tumor Pathology, Karolinska Institute, and Karolinska University Hospital, 17164 Stockholm, Sweden.
| |
Collapse
|
49
|
Manfredi LH, Ang J, Peker N, Dagda RK, McFarlane C. G protein-coupled receptor kinase 2 regulates mitochondrial bioenergetics and impairs myostatin-mediated autophagy in muscle cells. Am J Physiol Cell Physiol 2019; 317:C674-C686. [PMID: 31268780 PMCID: PMC6850988 DOI: 10.1152/ajpcell.00516.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 06/13/2019] [Accepted: 07/02/2019] [Indexed: 12/25/2022]
Abstract
G protein-coupled receptor kinase 2 (GRK2) is an important protein involved in β-adrenergic receptor desensitization. In addition, studies have shown GRK2 can modulate different metabolic processes in the cell. For instance, GRK2 has been recently shown to promote mitochondrial biogenesis and increase ATP production. However, the role of GRK2 in skeletal muscle and the signaling mechanisms that regulate GRK2 remain poorly understood. Myostatin is a well-known myokine that has been shown to impair mitochondria function. Here, we have assessed the role of myostatin in regulating GRK2 and the subsequent downstream effect of myostatin regulation of GRK2 on mitochondrial respiration in skeletal muscle. Myostatin treatment promoted the loss of GRK2 protein in myoblasts and myotubes in a time- and dose-dependent manner, which we suggest was through enhanced ubiquitin-mediated protein loss, as treatment with proteasome inhibitors partially rescued myostatin-mediated loss of GRK2 protein. To evaluate the effects of GRK2 on mitochondrial respiration, we generated stable myoblast lines that overexpress GRK2. Stable overexpression of GRK2 resulted in increased mitochondrial content and enhanced mitochondrial/oxidative respiration. Interestingly, although overexpression of GRK2 was unable to prevent myostatin-mediated impairment of mitochondrial respiratory function, elevated levels of GRK2 blocked the increased autophagic flux observed following treatment with myostatin. Overall, our data suggest a novel role for GRK2 in regulating mitochondria mass and mitochondrial respiration in skeletal muscle.
Collapse
MESH Headings
- Animals
- Autophagy/drug effects
- G-Protein-Coupled Receptor Kinase 2/drug effects
- G-Protein-Coupled Receptor Kinase 2/metabolism
- Mice
- Mitochondria/drug effects
- Mitochondria/metabolism
- Muscle Cells/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Myoblasts/drug effects
- Myoblasts/metabolism
- Myostatin/metabolism
- Myostatin/pharmacology
- Receptors, Adrenergic, beta/drug effects
- Receptors, Adrenergic, beta/metabolism
- Receptors, Adrenergic, beta-2/drug effects
- Receptors, Adrenergic, beta-2/metabolism
- Signal Transduction/drug effects
- Signal Transduction/physiology
Collapse
Affiliation(s)
- Leandro Henrique Manfredi
- Department of Physiology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- Federal University of Fronteira Sul, Medical School, Chapecó, Santa Catarina, Brazil
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research (A*STAR), Brenner Centre for Molecular Medicine, Singapore
| | - Joshur Ang
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research (A*STAR), Brenner Centre for Molecular Medicine, Singapore
| | - Nesibe Peker
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Ruben K Dagda
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, Nevada
| | - Craig McFarlane
- Singapore Institute for Clinical Sciences, Agency for Science, Technology, and Research (A*STAR), Brenner Centre for Molecular Medicine, Singapore
- Department of Molecular and Cell Biology, College of Public Health, Medical, and Veterinary Sciences, James Cook University, Townsville, Queensland, Australia
| |
Collapse
|
50
|
Kimura I, Ichimura A, Ohue-Kitano R, Igarashi M. Free Fatty Acid Receptors in Health and Disease. Physiol Rev 2019; 100:171-210. [PMID: 31487233 DOI: 10.1152/physrev.00041.2018] [Citation(s) in RCA: 488] [Impact Index Per Article: 97.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fatty acids are metabolized and synthesized as energy substrates during biological responses. Long- and medium-chain fatty acids derived mainly from dietary triglycerides, and short-chain fatty acids (SCFAs) produced by gut microbial fermentation of the otherwise indigestible dietary fiber, constitute the major sources of free fatty acids (FFAs) in the metabolic network. Recently, increasing evidence indicates that FFAs serve not only as energy sources but also as natural ligands for a group of orphan G protein-coupled receptors (GPCRs) termed free fatty acid receptors (FFARs), essentially intertwining metabolism and immunity in multiple ways, such as via inflammation regulation and secretion of peptide hormones. To date, several FFARs that are activated by the FFAs of various chain lengths have been identified and characterized. In particular, FFAR1 (GPR40) and FFAR4 (GPR120) are activated by long-chain saturated and unsaturated fatty acids, while FFAR3 (GPR41) and FFAR2 (GPR43) are activated by SCFAs, mainly acetate, butyrate, and propionate. In this review, we discuss the recent reports on the key physiological functions of the FFAR-mediated signaling transduction pathways in the regulation of metabolism and immune responses. We also attempt to reveal future research opportunities for developing therapeutics for metabolic and immune disorders.
Collapse
Affiliation(s)
- Ikuo Kimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| | - Atsuhiko Ichimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| | - Ryuji Ohue-Kitano
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| | - Miki Igarashi
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| |
Collapse
|