1
|
Dwyer MB, Aumiller JL, Wedegaertner PB. Going Rogue: Mechanisms, Regulation, and Roles of Mutationally Activated G α in Human Cancer. Mol Pharmacol 2024; 106:198-215. [PMID: 39187387 PMCID: PMC11493338 DOI: 10.1124/molpharm.124.000743] [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: 08/01/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 08/28/2024] Open
Abstract
G protein-coupled receptors (GPCRs) couple to heterotrimeric G proteins, comprised of α and βγ subunits, to convert extracellular signals into activation of intracellular signaling pathways. Canonically, GPCR-mediated activation results in the exchange of GDP for GTP on G protein α subunits (Gα) and the dissociation of Gα-GTP and G protein βγ subunits (Gβγ), both of which can regulate a variety of signaling pathways. Hydrolysis of bound GTP by Gα returns the protein to Gα-GDP and allows reassociation with Gβγ to reform the inactive heterotrimer. Naturally occurring mutations in Gα have been found at conserved glutamine and arginine amino acids that disrupt the canonical G protein cycle by inhibiting GTP hydrolysis, rendering these mutants constitutively active. Interestingly, these dysregulated Gα mutants are found in many different cancers due to their ability to sustain aberrant signaling without a need for activation by GPCRs. This review will highlight an increased recognition of the prevalence of such constitutively activating Gα mutations in cancers and the signaling pathways activated. In addition, we will discuss new knowledge regarding how these constitutively active Gα are regulated, how different mutations are biochemically distinct, and how mutationally activated Gα are unique compared with GPCR-activated Gα Lastly, we will discuss recent progress in developing inhibitors directly targeting constitutively active Gα mutants. SIGNIFICANCE STATEMENT: Constitutively activating mutations in G protein α subunits (Gα) widely occur in and contribute to the development of many human cancers. To develop ways to inhibit dysregulated, oncogenic signaling by these mutant Gα, it is crucial to better understand mechanisms that lead to constitutive Gα activation and unique mechanisms that regulate mutationally activated Gα in cells. The prevalence of activating mutations in Gα in various cancers makes Gα proteins compelling targets for the development of therapeutics.
Collapse
Affiliation(s)
- Morgan B Dwyer
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jenna L Aumiller
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Philip B Wedegaertner
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
2
|
Brands J, Bravo S, Jürgenliemke L, Grätz L, Schihada H, Frechen F, Alenfelder J, Pfeil C, Ohse PG, Hiratsuka S, Kawakami K, Schmacke LC, Heycke N, Inoue A, König G, Pfeifer A, Wachten D, Schulte G, Steinmetzer T, Watts VJ, Gomeza J, Simon K, Kostenis E. A molecular mechanism to diversify Ca 2+ signaling downstream of Gs protein-coupled receptors. Nat Commun 2024; 15:7684. [PMID: 39227390 PMCID: PMC11372221 DOI: 10.1038/s41467-024-51991-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024] Open
Abstract
A long-held tenet in inositol-lipid signaling is that cleavage of membrane phosphoinositides by phospholipase Cβ (PLCβ) isozymes to increase cytosolic Ca2+ in living cells is exclusive to Gq- and Gi-sensitive G protein-coupled receptors (GPCRs). Here we extend this central tenet and show that Gs-GPCRs also partake in inositol-lipid signaling and thereby increase cytosolic Ca2+. By combining CRISPR/Cas9 genome editing to delete Gαs, the adenylyl cyclase isoforms 3 and 6, or the PLCβ1-4 isozymes, with pharmacological and genetic inhibition of Gq and G11, we pin down Gs-derived Gβγ as driver of a PLCβ2/3-mediated cytosolic Ca2+ release module. This module does not require but crosstalks with Gαs-dependent cAMP, demands Gαq to release PLCβ3 autoinhibition, but becomes Gq-independent with mutational disruption of the PLCβ3 autoinhibited state. Our findings uncover the key steps of a previously unappreciated mechanism utilized by mammalian cells to finetune their calcium signaling regulation through Gs-GPCRs.
Collapse
Affiliation(s)
- Julian Brands
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
- Research Training Group 1873, University of Bonn, Bonn, Germany
| | - Sergi Bravo
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Lars Jürgenliemke
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
- Research Training Group 2873, University of Bonn, Bonn, Germany
| | - Lukas Grätz
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Hannes Schihada
- Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Fabian Frechen
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Judith Alenfelder
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Cy Pfeil
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
- Research Training Group 1873, University of Bonn, Bonn, Germany
- Amsterdam Institute for Molecular and Life Sciences (AIMMS), Division of Medicinal Chemistry, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Paul Georg Ohse
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Suzune Hiratsuka
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578, Japan
| | - Kouki Kawakami
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578, Japan
- Komaba Institute for Science, The University of Tokyo, Meguro, Tokyo, 153-8505, Japan
| | - Luna C Schmacke
- Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Nina Heycke
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, 980-8578, Japan
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Gabriele König
- Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Alexander Pfeifer
- Institute of Pharmacology and Toxicology, University Hospital, University of Bonn, Bonn, Germany
| | - Dagmar Wachten
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Gunnar Schulte
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Torsten Steinmetzer
- Department of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Val J Watts
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute of Drug Discovery, Purdue University, West Lafayette, IN, USA
| | - Jesús Gomeza
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Katharina Simon
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany.
| |
Collapse
|
3
|
Mastos C, Xu X, Keen AC, Halls ML. Signalling of Adrenoceptors: Canonical Pathways and New Paradigms. Handb Exp Pharmacol 2024; 285:147-184. [PMID: 38227198 DOI: 10.1007/164_2023_704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The concept of G protein-coupled receptors initially arose from studies of the β-adrenoceptor, adenylyl cyclase, and cAMP signalling pathway. Since then both canonical G protein-coupled receptor signalling pathways and emerging paradigms in receptor signalling have been defined by experiments focused on adrenoceptors. Here, we discuss the evidence for G protein coupling specificity of the nine adrenoceptor subtypes. We summarise the ability of each of the adrenoceptors to activate proximal signalling mediators including cAMP, calcium, mitogen-activated protein kinases, and protein kinase C pathways. Finally, we highlight the importance of precise spatial and temporal control of adrenoceptor signalling that is controlled by the localisation of receptors at intracellular membranes and in larger protein complexes.
Collapse
Affiliation(s)
- Chantel Mastos
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Xiaomeng Xu
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Alastair C Keen
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Michelle L Halls
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.
| |
Collapse
|
4
|
Sato K, Yamashita T, Ohuchi H. Mammalian type opsin 5 preferentially activates G14 in Gq-type G proteins triggering intracellular calcium response. J Biol Chem 2023; 299:105020. [PMID: 37423300 PMCID: PMC10432815 DOI: 10.1016/j.jbc.2023.105020] [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: 12/20/2022] [Revised: 06/28/2023] [Accepted: 07/01/2023] [Indexed: 07/11/2023] Open
Abstract
Mammalian type opsin 5 (Opn5m), a UV-sensitive G protein-coupled receptor opsin highly conserved in vertebrates, would provide a common basis for UV sensing from lamprey to humans. However, G protein coupled with Opn5m remains controversial due to variations in assay conditions and the origin of Opn5m across different reports. Here, we examined Opn5m from diverse species using an aequorin luminescence assay and Gα-KO cell line. Beyond the commonly studied major Gα classes, Gαq, Gα11, Gα14, and Gα15 in the Gq class were individually investigated in this study, as they can drive distinct signaling pathways in addition to a canonical calcium response. UV light triggered a calcium response via all the tested Opn5m proteins in 293T cells, which was abolished by Gq-type Gα deletion and rescued by cotransfection with mouse and medaka Gq-type Gα proteins. Opn5m preferentially activated Gα14 and close relatives. Mutational analysis implicated specific regions, including α3-β5 and αG-α4 loops, αG and α4 helices, and the extreme C terminus, in the preferential activation of Gα14 by Opn5m. FISH revealed co-expression of genes encoding Opn5m and Gα14 in the scleral cartilage of medaka and chicken eyes, supporting their physiological coupling. This suggests that the preferential activation of Gα14 by Opn5m is relevant for UV sensing in specific cell types.
Collapse
Affiliation(s)
- Keita Sato
- Department of Cytology and Histology, Faculty of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama City, Okayama, Japan.
| | - Takahiro Yamashita
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto, Japan
| | - Hideyo Ohuchi
- Department of Cytology and Histology, Faculty of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama City, Okayama, Japan.
| |
Collapse
|
5
|
Kulkarni VV, Wang Y, Pantaleon Garcia J, Evans SE. Redox-Dependent Activation of Lung Epithelial STAT3 Is Required for Inducible Protection against Bacterial Pneumonia. Am J Respir Cell Mol Biol 2023; 68:679-688. [PMID: 36826841 PMCID: PMC10257071 DOI: 10.1165/rcmb.2022-0342oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 02/24/2023] [Indexed: 02/25/2023] Open
Abstract
The lung epithelium is dynamic, capable of considerable structural and functional plasticity in response to pathogen challenges. Our laboratory has demonstrated that an inhaled combination of a Toll-like receptor (TLR) 2/6 agonist and a TLR9 agonist (Pam2ODN) results in robust protection against otherwise lethal pneumonias. We have previously shown that intact epithelial TLR signaling and generation of multisource epithelial reactive oxygen species (ROS) are required for inducible protection. Further investigating the mechanisms underlying this phenomenon of inducible resistance, reverse-phase protein array analysis demonstrated robust STAT3 (signal transducer and activator of transcription 3) phosphorylation following treatment of lung epithelial cells. We show here that Pam2ODN-induced STAT3 phosphorylation is IL-6-independent. We further found that therapeutic epithelial STAT3 activation is required for inducible protection against Pseudomonas aeruginosa pneumonia. Additional studies showed that inhibiting epithelial dual oxidases or scavenging ROS significantly reduced the Pam2ODN induction of STAT3 phosphorylation, suggesting a proximal role for ROS in inducible STAT3 activation. Dissecting these mechanisms, we analyzed the contributions of redox-sensitive kinases and found that Pam2ODN activated epithelial growth factor receptor in an ROS-dependent manner that is required for therapeutically inducible STAT3 activation. Taken together, we demonstrate that epithelial STAT3 is imperative for Pam2ODN's function and describe a novel redox-based mechanism for its activation. These key mechanistic insights may facilitate strategies to leverage inducible epithelial resistance to protect susceptible patients during periods of peak vulnerability.
Collapse
Affiliation(s)
- Vikram V. Kulkarni
- MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, Texas; and
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yongxing Wang
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Scott E. Evans
- MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, Texas; and
- Department of Pulmonary Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
6
|
Imaging of Gα q Proteins in Mouse and Human Organs and Tissues. Pharmaceutics 2022; 15:pharmaceutics15010057. [PMID: 36678686 PMCID: PMC9865079 DOI: 10.3390/pharmaceutics15010057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
G protein-coupled receptors (GPCRs) transfer extracellular signals across cell membranes by activating intracellular heterotrimeric G proteins. Several studies suggested G proteins as novel drug targets for the treatment of complex diseases, e.g., asthma and cancer. Recently, we developed specific radiotracers, [³H]PSB-15900-FR and [³H]PSB-16254-YM, for the Gαq family of G proteins by tritiation of the macrocyclic natural products FR900359 (FR) and YM-254890 (YM). In the present study, we utilized these potent radioligands to perform autoradiography studies in tissues of healthy mice, mouse models of disease, and human tissues. Specific binding was high, while non-specific binding was extraordinarily low, giving nearly identical results for both radioligands. High expression levels of Gαq proteins were detected in healthy mouse organs showing the following rank order of potency: kidney > liver > brain > pancreas > lung > spleen, while expression in the heart was low. Organ sub-structures, e.g., of mouse brain and lung, were clearly distinguishable. Whereas an acute asthma model in mice did not result in altered Gαq protein expressions as compared to control animals, a cutaneous melanoma model displayed significantly increased expression in comparison to healthy skin. These results suggest the future development of Gαq-protein-binding radio-tracers as novel diagnostics.
Collapse
|
7
|
Katsouri IP, Vandervelpen EVG, Gattor AO, Engelbeen S, El Sayed A, Seitaj K, Becerra EDM, Vanderheyden PML. Complex FFA1 receptor (in)dependent modulation of calcium signaling by free fatty acids. Biochem Pharmacol 2022; 202:115150. [PMID: 35724691 DOI: 10.1016/j.bcp.2022.115150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 11/26/2022]
Abstract
The expression of free fatty acid 1 receptors (FFA1R), activated by long chain fatty acids in human pancreatic β-cells and enhancing glucose-stimulated insulin secretion are an attractive target to treat type 2 diabetes. Yet several clinical studies with synthetic FFA1R agonists had to be discontinued due to cytotoxicity and/or so-called "liver concerns". It is not clear whether these obstructions are FFA1R dependent. In this context we used CHO-AEQ cells expressing the bioluminescent calcium-sensitive protein aequorin to investigate calcium signaling elicited by FFA1 receptor ligands α-linolenic acid (ALA), oleic acid (OLA) and myristic acid (MYA). This study revealed complex modulation of intracellular calcium signaling by these fatty acids. First these compounds elicited a typical transient increase of intracellular calcium via binding to FFA1 receptors. Secondly slightly higher concentrations of ALA substantially reduced ATP mediated calcium responses in CHO-AEQ cells and Angiotensin II responses in CHO-AEQ cells expressing human AT1 receptors. This effect was less pronounced with MYA and OLA and was not linked to FFA1 receptor activation nor to acute cytotoxicity as a result of plasma membrane perturbation. Yet it can be hypothesized that, in line with previous studies, unsaturated long chain fatty acids such as ALA and OLA are capable of inactivating the G-proteins involved in purinergic and Angiotensin AT1 receptor calcium signaling. Alternatively the ability of fatty acids to deplete intracellular calcium stores might underly the observed cross-inhibition of these receptor responses in the same cells.
Collapse
Affiliation(s)
- Ilektra Petrina Katsouri
- Research Group of Molecular and Biochemical Pharmacology, Department of Biotechnology and Bioengineering, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ebert Vinciane G Vandervelpen
- Research Group of Molecular and Biochemical Pharmacology, Department of Biotechnology and Bioengineering, Vrije Universiteit Brussel, Brussels, Belgium
| | - Albert Owusu Gattor
- Lehrstuhl für Pharmazeutische und Medizinische Chemie II, Universität Regensburg, Regensburg, Germany
| | - Sarah Engelbeen
- Research Group of Molecular and Biochemical Pharmacology, Department of Biotechnology and Bioengineering, Vrije Universiteit Brussel, Brussels, Belgium
| | - Abdulrahman El Sayed
- The International Institute of Molecular Mechanisms and Machines, Polish Academy of Sciences, Warsaw, Poland
| | - Klejdia Seitaj
- Research Group of Molecular and Biochemical Pharmacology, Department of Biotechnology and Bioengineering, Vrije Universiteit Brussel, Brussels, Belgium
| | - Eduardo Daniel Morales Becerra
- Research Group of Molecular and Biochemical Pharmacology, Department of Biotechnology and Bioengineering, Vrije Universiteit Brussel, Brussels, Belgium
| | - Patrick M L Vanderheyden
- Research Group of Molecular and Biochemical Pharmacology, Department of Biotechnology and Bioengineering, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
8
|
Chan KYY, Chung PY, Zhang C, Poon ENY, Leung AWK, Leung KT. R4 RGS proteins as fine tuners of immature and mature hematopoietic cell trafficking. J Leukoc Biol 2022; 112:785-797. [PMID: 35694792 DOI: 10.1002/jlb.1mr0422-475r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/28/2022] [Indexed: 11/08/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) are the largest and most diverse group of membrane receptors. They are involved in almost every physiologic process and consequently have a pivotal role in an extensive number of pathologies, including genetic, neurologic, and immune system disorders. Indeed, the vast array of GPCRs mechanisms have led to the development of a tremendous number of drug therapies and already account for about a third of marketed drugs. These receptors mediate their downstream signals primarily via G proteins. The regulators of G-protein signaling (RGS) proteins are now in the spotlight as the critical modulatory factors of active GTP-bound Gα subunits of heterotrimeric G proteins to fine-tune the biologic responses driven by the GPCRs. Also, they possess noncanonical functions by multiple mechanisms, such as protein-protein interactions. Essential roles and impacts of these RGS proteins have been revealed in physiology, including hematopoiesis and immunity, and pathologies, including asthma, cancers, and neurologic disorders. This review focuses on the largest subfamily of R4 RGS proteins and provides a brief overview of their structures and G-proteins selectivity. With particular interest, we explore and highlight, their expression in the hematopoietic system and the regulation in the engraftment of hematopoietic stem/progenitor cells (HSPCs). Distinct expression patterns of R4 RGS proteins in the hematopoietic system and their pivotal roles in stem cell trafficking pave the way for realizing new strategies for enhancing the clinical performance of hematopoietic stem cell transplantation. Finally, we discuss the exciting future trends in drug development by targeting RGS activity and expression with small molecules inhibitors and miRNA approaches.
Collapse
Affiliation(s)
- Kathy Yuen Yee Chan
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Po Yee Chung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Chi Zhang
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Ellen Ngar Yun Poon
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Alex Wing Kwan Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong, China.,Department of Paediatrics & Adolescent Medicine, Hong Kong Children's Hospital, Hong Kong SAR, China
| | - Kam Tong Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong, China.,Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| |
Collapse
|
9
|
Voss JH, Mahardhika AB, Inoue A, Müller CE. Agonist-Dependent Coupling of the Promiscuous Adenosine A 2B Receptor to Gα Protein Subunits. ACS Pharmacol Transl Sci 2022; 5:373-386. [PMID: 35592437 PMCID: PMC9112290 DOI: 10.1021/acsptsci.2c00020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Indexed: 12/28/2022]
Abstract
The adenosine A2B receptor (A2BAR) belongs to the rhodopsin-like G protein-coupled receptor (GPCR) family. It is upregulated under hypoxic conditions, in inflammation and cancer. Previous studies indicated the coupling of the A2BAR to different G proteins, mainly Gs, but in some cases Gq/11 or Gi, depending on the cell type. We have now utilized novel technologies, (i) heterologous expression of individual members of the Gαq/11 protein family (Gαq, Gα11, Gα14, and Gα15) in Gαq/11 knockout cells, and (ii) the TRUPATH platform, allowing the direct observation of Gα protein activation for each of the Gα subunits by bioluminescence resonance energy transfer (BRET) measurements. Three structurally diverse A2BAR agonists were studied: the cognate agonist adenosine, its metabolically stable analog NECA, and the non-nucleosidic partial agonist BAY 60-6583. Adenosine and NECA activated most members of all four Gα protein families (Gαs, Gαq/11, Gαi, and Gα12/13). Significant differences in potencies and efficacies were observed; the highest efficacies were determined at the Gα15, Gαs, and Gα12 proteins, and for NECA additionally at the Gαi2 protein. In contrast, the partial agonist BAY 60-6583 only activated Gα15, Gαs, and Gα12 proteins. Adenosine deaminase, an allosteric modulator of ARs, selectively increased the potency and efficacy of NECA and BAY 60-6583 at the Gα15 protein, while it had no effect or decreased efficacy at the other Gα proteins. We conclude that the A2BAR is preferably coupled to the Gα15, Gαs, and Gα12 proteins. Upon upregulation of receptor or Gα protein expression, coupling to further Gα proteins likely occurs. Importantly, different agonists can display different activation profiles.
Collapse
Affiliation(s)
- Jan Hendrik Voss
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Andhika B Mahardhika
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany.,Research Training Group GRK1873, University of Bonn, D-53121 Bonn, Germany
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany.,Research Training Group GRK1873, University of Bonn, D-53121 Bonn, Germany
| |
Collapse
|
10
|
Sánchez Triviño CA, Landinez MP, Duran S, Gomez MDP, Nasi E. Modulation of Gq/PLC-Mediated Signaling by Acute Lithium Exposure. Front Cell Neurosci 2022; 16:838939. [PMID: 35242014 PMCID: PMC8885521 DOI: 10.3389/fncel.2022.838939] [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: 12/18/2021] [Accepted: 01/27/2022] [Indexed: 11/13/2022] Open
Abstract
Although lithium has long been one of the most widely used pharmacological agents in psychiatry, its mechanisms of action at the cellular and molecular levels remain poorly understood. One of the targets of Li+ is the phosphoinositide pathway, but whereas the impact of Li+ on inositol lipid metabolism is well documented, information on physiological effects at the cellular level is lacking. We examined in two mammalian cell lines the effect of acute Li+ exposure on the mobilization of internal Ca2+ and phospholipase C (PLC)-dependent membrane conductances. We first corroborated by Western blots and immunofluorescence in HEK293 cells the presence of key signaling elements of a muscarinic PLC pathway (M1AchR, Gq, PLC-β1, and IP3Rs). Stimulation with carbachol evoked a dose-dependent mobilization of Ca, as determined with fluorescent indicators. This was due to release from internal stores and proved susceptible to the PLC antagonist U73122. Li+ exposure reproducibly potentiated the Ca response in a concentration-dependent manner extending to the low millimolar range. To broaden those observations to a neuronal context and probe potential Li modulation of electrical signaling, we next examined the cell line SHsy5y. We replicated the potentiating effects of Li on the mobilization of internal Ca, and, after characterizing the basic properties of the electrical response to cholinergic stimulation, we also demonstrated an equally robust upregulation of muscarinic membrane currents. Finally, by directly stimulating the signaling pathway at different links downstream of the receptor, the site of action of the observed Li effects could be narrowed down to the G protein and its interaction with PLC-β. These observations document a modulation of Gq/PLC/IP3-mediated signaling by acute exposure to lithium, reflected in distinct physiological changes in cellular responses.
Collapse
Affiliation(s)
- Cesar Adolfo Sánchez Triviño
- Departamento de Biología, Universidad Nacional de Colombia, Bogotá, Colombia
- Centro Internacional de Física, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Maria Paula Landinez
- Departamento de Biología, Universidad Nacional de Colombia, Bogotá, Colombia
- Centro Internacional de Física, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Sara Duran
- Departamento de Biología, Universidad Nacional de Colombia, Bogotá, Colombia
- Centro Internacional de Física, Universidad Nacional de Colombia, Bogotá, Colombia
| | - María Del Pilar Gomez
- Departamento de Biología, Universidad Nacional de Colombia, Bogotá, Colombia
- Marine Biological Laboratory, Woods Hole, MA, United States
| | - Enrico Nasi
- Marine Biological Laboratory, Woods Hole, MA, United States
- Instituto de Genética, Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
11
|
Li M, Liu Y, Liu Y, Yang L, Xu Y, Wang W, Jiang Z, Liu Y, Wang S, Wang C. Downregulation of GNA15 Inhibits Cell Proliferation via P38 MAPK Pathway and Correlates with Prognosis of Adult Acute Myeloid Leukemia With Normal Karyotype. Front Oncol 2021; 11:724435. [PMID: 34552875 PMCID: PMC8451478 DOI: 10.3389/fonc.2021.724435] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/17/2021] [Indexed: 11/13/2022] Open
Abstract
Background The prognosis of acute myeloid leukemia (AML) with a normal karyotype is highly heterogonous, and the current risk stratification is still insufficient to differentiate patients from high-risk to standard-risk. Changes in some genetic profiles may contribute to the poor prognosis of AML. Although the prognostic value of G protein subunit alpha 15 (GNA15) in AML has been reported based on the GEO (Gene Expression Omnibus) database, the prognostic significance of GNA15 has not been verified in clinical samples. The biological functions of GNA15 in AML development remain open to investigation. This study explored the clinical significance, biological effects and molecular mechanism of GNA15 in AML. Methods Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to detect the mRNA expression level of GNA15 in blasts of bone marrow specimens from 154 newly diagnosed adult AML patients and 26 healthy volunteers. AML cell lines, Kasumi-1 and SKNO-1, were used for lentiviral transfection. Cell Counting Kit-8 (CCK8) and colony formation assays were used to determine cell proliferation. Cell cycle and apoptosis were analyzed by flow cytometry. The relevant signaling pathways were evaluated by Western blot. The Log-Rank test and Kaplan-Meier were used to evaluate survival rate, and the Cox regression model was used to analyze multivariate analysis. Xenograft tumor mouse model was used for in vivo experiments. Results The expression of GNA15 in adult AML was significantly higher than that in healthy individuals. Subjects with high GNA15 expression showed lower overall survival and relapse-free survival in adult AML with normal karyotype. High GNA15 expression was independently correlated with a worse prognosis in multivariate analysis. Knockdown of GNA15 inhibited cell proliferation and cell cycle progression, and induced cell apoptosis in AML cells. GNA15-knockdown induced down-regulation of p-P38 MAPK and its downstream p-MAPKAPK2 and p-CREB. Rescue assays confirmed that P38 MAPK signaling pathway was involved in the inhibition of proliferation mediated by GNA15 knockdown. Conclusions In summary, GNA15 was highly expressed in adult AML, and high GNA15 expression was independently correlated with a worse prognosis in adult AML with normal karyotype. Knockdown of GNA15 inhibited the proliferation of AML regulated by the P38 MAPK signaling pathway. Therefore, GNA15 may serve as a potential prognostic marker and a therapeutic target for AML in the future.
Collapse
Affiliation(s)
- Mengya Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yu Liu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yajun Liu
- Department of Orthopaedics, Rhode Island Hospital, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | - Lu Yang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Xu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weiqiong Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanfang Liu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shujuan Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chong Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
12
|
Fusion with Promiscuous Gα 16 Subunit Reveals Signaling Bias at Muscarinic Receptors. Int J Mol Sci 2021; 22:ijms221810089. [PMID: 34576254 PMCID: PMC8469978 DOI: 10.3390/ijms221810089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/06/2021] [Accepted: 09/14/2021] [Indexed: 11/25/2022] Open
Abstract
A complex evaluation of agonist bias at G-protein coupled receptors at the level of G-protein classes and isoforms including non-preferential ones is essential for advanced agonist screening and drug development. Molecular crosstalk in downstream signaling and a lack of sufficiently sensitive and selective methods to study direct coupling with G-protein of interest complicates this analysis. We performed binding and functional analysis of 11 structurally different agonists on prepared fusion proteins of individual subtypes of muscarinic receptors and non-canonical promiscuous α-subunit of G16 protein to study agonist bias. We have demonstrated that fusion of muscarinic receptors with Gα16 limits access of other competitive Gα subunits to the receptor, and thus enables us to study activation of Gα16 mediated pathway more specifically. Our data demonstrated agonist-specific activation of G16 pathway among individual subtypes of muscarinic receptors and revealed signaling bias of oxotremorine towards Gα16 pathway at the M2 receptor and at the same time impaired Gα16 signaling of iperoxo at M5 receptors. Our data have shown that fusion proteins of muscarinic receptors with α-subunit of G-proteins can serve as a suitable tool for studying agonist bias, especially at non-preferential pathways.
Collapse
|
13
|
Tripolt S, Neubauer HA, Knab VM, Elmer DP, Aberger F, Moriggl R, Fux DA. Opioids drive breast cancer metastasis through the δ-opioid receptor and oncogenic STAT3. Neoplasia 2021; 23:270-279. [PMID: 33465556 PMCID: PMC7815495 DOI: 10.1016/j.neo.2020.12.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 12/25/2020] [Accepted: 12/29/2020] [Indexed: 11/25/2022]
Abstract
The opioid crisis of pain medication bears risks from addiction to cancer progression, but little experimental evidence exists. Expression of δ-opioid receptors (DORs) correlates with poor prognosis for breast cancer patients, but mechanistic insights into oncogenic signaling mechanisms of opioid-triggered cancer progression are lacking. We show that orthotopic transplant models using human or murine breast cancer cells displayed enhanced metastasis upon opioid-induced DOR stimulation. Interestingly, opioid-exposed breast cancer cells showed enhanced migration and strong STAT3 activation, which was efficiently blocked by a DOR-antagonist. Furthermore, opioid treatment resulted in down-regulation of E-Cadherin and increased expression of epithelial-mesenchymal transition markers. Notably, STAT3 knockdown or upstream inhibition through the JAK1/2 kinase inhibitor ruxolitinib prevented opioid-induced breast cancer cell metastasis and migration in vitro and in vivo. We conclude on a novel mechanism whereby opioid-triggered breast cancer metastasis occurs via oncogenic JAK1/2-STAT3 signaling to promote epithelial-mesenchymal transition. These findings emphasize the importance of selective and restricted opioid use, as well as the need for safer pain medication that does not activate these oncogenic pathways.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Biomarkers
- Breast Neoplasms/etiology
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Cell Line, Tumor
- Disease Models, Animal
- Disease Susceptibility
- Female
- Gene Expression
- Humans
- Immunohistochemistry
- Mice
- Mice, Transgenic
- Neoplasm Metastasis
- Oncogene Proteins/metabolism
- Receptors, Opioid, delta/agonists
- Receptors, Opioid, delta/genetics
- Receptors, Opioid, delta/metabolism
- STAT3 Transcription Factor/metabolism
Collapse
Affiliation(s)
- Sabrina Tripolt
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Heidi A Neubauer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Vanessa M Knab
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Dominik P Elmer
- Department of Biosciences, Cancer Cluster Salzburg, Paris-Lodron University of Salzburg, Salzburg, Austria
| | - Fritz Aberger
- Department of Biosciences, Cancer Cluster Salzburg, Paris-Lodron University of Salzburg, Salzburg, Austria
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Daniela A Fux
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine Vienna, Vienna, Austria.
| |
Collapse
|
14
|
Spanoghe J, Larsen LE, Craey E, Manzella S, Van Dycke A, Boon P, Raedt R. The Signaling Pathways Involved in the Anticonvulsive Effects of the Adenosine A 1 Receptor. Int J Mol Sci 2020; 22:ijms22010320. [PMID: 33396826 PMCID: PMC7794785 DOI: 10.3390/ijms22010320] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/22/2020] [Accepted: 12/27/2020] [Indexed: 12/20/2022] Open
Abstract
Adenosine acts as an endogenous anticonvulsant and seizure terminator in the brain. Many of its anticonvulsive effects are mediated through the activation of the adenosine A1 receptor, a G protein-coupled receptor with a wide array of targets. Activating A1 receptors is an effective approach to suppress seizures. This review gives an overview of the neuronal targets of the adenosine A1 receptor focusing in particular on signaling pathways resulting in neuronal inhibition. These include direct interactions of G protein subunits, the adenyl cyclase pathway and the phospholipase C pathway, which all mediate neuronal hyperpolarization and suppression of synaptic transmission. Additionally, the contribution of the guanyl cyclase and mitogen-activated protein kinase cascades to the seizure-suppressing effects of A1 receptor activation are discussed. This review ends with the cautionary note that chronic activation of the A1 receptor might have detrimental effects, which will need to be avoided when pursuing A1 receptor-based epilepsy therapies.
Collapse
Affiliation(s)
- Jeroen Spanoghe
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium; (J.S.); (L.E.L.); (E.C.); (S.M.); (P.B.)
| | - Lars E. Larsen
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium; (J.S.); (L.E.L.); (E.C.); (S.M.); (P.B.)
| | - Erine Craey
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium; (J.S.); (L.E.L.); (E.C.); (S.M.); (P.B.)
| | - Simona Manzella
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium; (J.S.); (L.E.L.); (E.C.); (S.M.); (P.B.)
| | - Annelies Van Dycke
- Department of Neurology, General Hospital Sint-Jan Bruges, 8000 Bruges, Belgium;
| | - Paul Boon
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium; (J.S.); (L.E.L.); (E.C.); (S.M.); (P.B.)
| | - Robrecht Raedt
- 4Brain, Department of Head and Skin, Ghent University, 9000 Ghent, Belgium; (J.S.); (L.E.L.); (E.C.); (S.M.); (P.B.)
- Correspondence:
| |
Collapse
|
15
|
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: 0.5] [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
|
16
|
Cecon E, Oishi A, Jockers R. Melatonin receptors: molecular pharmacology and signalling in the context of system bias. Br J Pharmacol 2018; 175:3263-3280. [PMID: 28707298 PMCID: PMC6057902 DOI: 10.1111/bph.13950] [Citation(s) in RCA: 173] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 07/05/2017] [Accepted: 07/10/2017] [Indexed: 12/15/2022] Open
Abstract
Melatonin, N-acetyl-5-methoxytryptamine, an evolutionally old molecule, is produced by the pineal gland in vertebrates, and it binds with high affinity to melatonin receptors, which are members of the GPCR family. Among the multiple effects attributed to melatonin, we will focus here on those that are dependent on the activation of the two mammalian MT1 and MT2 melatonin receptors. We briefly summarize the latest developments on synthetic melatonin receptor ligands, including multi-target-directed ligands, and the characterization of signalling-biased ligands. We discuss signalling pathways activated by melatonin receptors that appear to be highly cell- and tissue-dependent, emphasizing the impact of system bias on the functional outcome. Different proteins have been demonstrated to interact with melatonin receptors, and thus, we postulate that part of this system bias has its molecular basis in differences of the expression of receptor-associated proteins including heterodimerization partners. Finally, bias at the level of the receptor, by the expression of genetic receptor variants, will be discussed to show how a modified receptor function can have an effect on the risk for common diseases like type 2 diabetes in humans. LINKED ARTICLES: This article is part of a themed section on Recent Developments in Research of Melatonin and its Potential Therapeutic Applications. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.16/issuetoc.
Collapse
Affiliation(s)
- Erika Cecon
- Institut CochinInserm, U1016ParisFrance
- CNRS UMR 8104ParisFrance
- Univ. Paris Descartes, Sorbonne Paris CitéParisFrance
| | - Atsuro Oishi
- Institut CochinInserm, U1016ParisFrance
- CNRS UMR 8104ParisFrance
- Univ. Paris Descartes, Sorbonne Paris CitéParisFrance
| | - Ralf Jockers
- Institut CochinInserm, U1016ParisFrance
- CNRS UMR 8104ParisFrance
- Univ. Paris Descartes, Sorbonne Paris CitéParisFrance
| |
Collapse
|
17
|
I KY, Huang YS, Hu CH, Tseng WY, Cheng CH, Stacey M, Gordon S, Chang GW, Lin HH. Activation of Adhesion GPCR EMR2/ADGRE2 Induces Macrophage Differentiation and Inflammatory Responses via Gα 16/Akt/MAPK/NF-κB Signaling Pathways. Front Immunol 2017; 8:373. [PMID: 28421075 PMCID: PMC5376562 DOI: 10.3389/fimmu.2017.00373] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/15/2017] [Indexed: 11/13/2022] Open
Abstract
EMR2/ADGRE2 is a human myeloid-restricted adhesion G protein-coupled receptor critically implicated in vibratory urticaria, a rare type of allergy caused by vibration-induced mast cell activation. In addition, EMR2 is also highly expressed by monocyte/macrophages and has been linked to neutrophil migration and activation. Despite these findings, little is known of EMR2-mediated signaling and its role in myeloid biology. In this report, we show that activation of EMR2 via a receptor-specific monoclonal antibody promotes the differentiation of human THP-1 monocytic cell line and induces the expression of pro-inflammatory mediators, including IL-8, TNF-α, and MMP-9. Using specific signaling inhibitors and siRNA knockdowns, biochemical and functional analyses reveal that the EMR2-mediated signaling is initiated by Gα16, followed by the subsequent activation of Akt, extracellular signal-regulated kinase, c-Jun N-terminal kinase, and nuclear factor kappa-light-chain-enhancer of activated B cells. Our results demonstrate a functional role for EMR2 in the differentiation and inflammatory activation of human monocytic cells and provide potential targets for myeloid cell-mediated inflammatory disorders.
Collapse
Affiliation(s)
- Kuan-Yu I
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Shu Huang
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ching-Hsun Hu
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wen-Yi Tseng
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital-Keelung, Keelung, Taiwan
| | - Chia-Hsin Cheng
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Martin Stacey
- Faculty of Biological Sciences, School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Siamon Gordon
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Gin-Wen Chang
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsi-Hsien Lin
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Anatomic Pathology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan.,Chang Gung Immunology Consortium, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
18
|
Koop G, Vrieling M, Storisteanu DML, Lok LSC, Monie T, van Wigcheren G, Raisen C, Ba X, Gleadall N, Hadjirin N, Timmerman AJ, Wagenaar JA, Klunder HM, Fitzgerald JR, Zadoks R, Paterson GK, Torres C, Waller AS, Loeffler A, Loncaric I, Hoet AE, Bergström K, De Martino L, Pomba C, de Lencastre H, Ben Slama K, Gharsa H, Richardson EJ, Chilvers ER, de Haas C, van Kessel K, van Strijp JAG, Harrison EM, Holmes MA. Identification of LukPQ, a novel, equid-adapted leukocidin of Staphylococcus aureus. Sci Rep 2017; 7:40660. [PMID: 28106142 PMCID: PMC5247767 DOI: 10.1038/srep40660] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/08/2016] [Indexed: 11/09/2022] Open
Abstract
Bicomponent pore-forming leukocidins are a family of potent toxins secreted by Staphylococcus aureus, which target white blood cells preferentially and consist of an S- and an F-component. The S-component recognizes a receptor on the host cell, enabling high-affinity binding to the cell surface, after which the toxins form a pore that penetrates the cell lipid bilayer. Until now, six different leukocidins have been described, some of which are host and cell specific. Here, we identify and characterise a novel S. aureus leukocidin; LukPQ. LukPQ is encoded on a 45 kb prophage (ΦSaeq1) found in six different clonal lineages, almost exclusively in strains cultured from equids. We show that LukPQ is a potent and specific killer of equine neutrophils and identify equine-CXCRA and CXCR2 as its target receptors. Although the S-component (LukP) is highly similar to the S-component of LukED, the species specificity of LukPQ and LukED differs. By forming non-canonical toxin pairs, we identify that the F-component contributes to the observed host tropism of LukPQ, thereby challenging the current paradigm that leukocidin specificity is driven solely by the S-component.
Collapse
Affiliation(s)
- Gerrit Koop
- Department of Farm Animal Health, Faculty of Veterinary Medicine, Utrecht University, 3584 CL, Utrecht, The Netherlands
| | - Manouk Vrieling
- Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Daniel M. L. Storisteanu
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s and Papworth Hospitals, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Laurence S. C. Lok
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s and Papworth Hospitals, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Tom Monie
- Medical Research Council Human Nutrition Research, Elsie Widdowson Laboratory, 120 Fulbourn Road, Cambridge CB1 9NL, United Kingdom
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| | - Glenn van Wigcheren
- Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Claire Raisen
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| | - Xiaoliang Ba
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| | - Nicholas Gleadall
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| | - Nazreen Hadjirin
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| | - Arjen J. Timmerman
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
| | - Jaap A. Wagenaar
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands
- Central Veterinary Institute of Wageningen UR, 8200 AB Lelystad, The Netherlands
| | - Heleen M. Klunder
- Department of Farm Animal Health, Faculty of Veterinary Medicine, Utrecht University, 3584 CL, Utrecht, The Netherlands
| | - J. Ross Fitzgerald
- The Roslin Institute, University of Edinburgh, EH25 9RG, Edinburgh, United Kingdom
| | - Ruth Zadoks
- Moredun Research Institute, Bush Loan, Penicuik EH26 0PZ, United Kingdom
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G61 1QH, United Kingdom
| | - Gavin K. Paterson
- Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG, United Kingdom
| | - Carmen Torres
- Área Bioquímica y Biología Molecular, Universidad de La Rioja, Madre de Dios 51, Logroño 26006, Spain
| | - Andrew S. Waller
- Animal Health Trust, Lanwades Park, Kentford, Newmarket CB8 7UU, United Kingdom
| | - Anette Loeffler
- Department of Clinical Sciences and Services, Royal Veterinary College, Hawkshead Lane, Hatfield, North Mymms, Hertfordshire AL9 7TA, United Kingdom
| | - Igor Loncaric
- Institute of Microbiology, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Armando E. Hoet
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, 1900 Coffey Road, Columbus, OH 43210, USA
- Veterinary Public Health Program, College of Public Health, The Ohio State University, 1900 Coffey Road, Columbus, OH 43210, USA
| | - Karin Bergström
- Department of Animal Health and Antimicrobial Strategies, SVA, SE-751 89 Uppsala, Sweden
| | - Luisa De Martino
- Department of Veterinary Medicine and Animal Production, Infectious Diseases Section, University of Naples “Federico II”, 80137 Naples, Italy
| | - Constança Pomba
- Interdisciplinary Centre of Research in Animal Health, Faculdade de Medicina Veterinária, Universidade de Lisboa, 1300-477 LISBOA, Portugal
| | - Hermínia de Lencastre
- Laboratório de Genética Molecular, Instituto de Tecnologia Química e Biológica da Universidade Nova de Lisboa (ITQB/UNL), Oeiras, Portugal
- Laboratory of Microbiology and Infectious Diseases, The Rockefeller University, New York, NY10065, USA
| | - Karim Ben Slama
- Laboratoire de Microorganismes et Biomolécules actives, Département de Biologie, Faculté de Sciences de Tunis, 2092 Tunis, Tunisia
- Institut Supérieur des Sciences Biologiques Appliquées de Tunis, Université de Tunis El Manar, 2092 Tunis, Tunisia
| | - Haythem Gharsa
- Laboratoire de Microorganismes et Biomolécules actives, Département de Biologie, Faculté de Sciences de Tunis, 2092 Tunis, Tunisia
| | - Emily J. Richardson
- Institute of Microbiology and Infection, University of Birmingham, Birmingham B15 2TT, UK
| | - Edwin R. Chilvers
- Department of Medicine, University of Cambridge School of Clinical Medicine, Addenbrooke’s and Papworth Hospitals, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Carla de Haas
- Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Kok van Kessel
- Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Jos A. G. van Strijp
- Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Ewan M. Harrison
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Mark A. Holmes
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| |
Collapse
|
19
|
GNAQ and GNA11 mutations occur in 9.5% of mucosal melanoma and are associated with poor prognosis. Eur J Cancer 2016; 65:156-63. [DOI: 10.1016/j.ejca.2016.06.019] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 06/02/2016] [Accepted: 06/20/2016] [Indexed: 12/22/2022]
|
20
|
Requirement of Gα q/Gα 11 Signaling in the Preservation of Mouse Intestinal Epithelial Homeostasis. Cell Mol Gastroenterol Hepatol 2016; 2:767-782.e6. [PMID: 28174748 PMCID: PMC5247319 DOI: 10.1016/j.jcmgh.2016.08.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 08/15/2016] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Proliferation, differentiation, and morphogenesis of the intestinal epithelium are tightly regulated by a number of molecular pathways. Coordinated action of intestine is achieved by gastrointestinal hormones, most of which exert these actions through G-protein-coupled receptors. We herein investigated the role of Gαq/11-mediated signaling in intestinal homeostasis. METHODS Intestinal tissues from control (Gnaqflox/floxGna11+/+ ), Int-Gq knock-out (KO) (VilCre+/-Gnaqflox/floxGna11+/+ ), G11 KO (Gnaqflox/floxGna11-/- ), and Int-Gq/G11 double knock-out (DKO) (VilCre+/-Gnaqflox/floxGna11-/- ) mice were examined by microscopy, transmission electron microscopy, and immunohistochemistry. The effect of Gαq/11-mediated signaling was studied in the cell lineage, proliferation, and apoptosis. Dextran sodium sulfate (DSS) colitis was induced to study the role of Gαq/11 in colon. RESULTS Paneth cells were enlarged, increased in number, and mislocalized in Int-Gq/G11 DKO small intestine. Paneth cells also reacted with PAS and Muc2 antibody, indicating an intermediate character of Paneth and goblet cells. The nuclear β-catenin, T-cell factor 1, and Sox9 expression were reduced severely in the crypt base of Int-Gq/G11 DKO intestine. Proliferation was activated in the crypt base and apoptosis was enhanced along the crypt. Int-Gq/G11 DKO mice were susceptible to DSS colitis. Proliferation was inhibited in the crypt of unaffected and regenerative areas. Cystic crypts, periodic acid-Schiff-positive cells, and Muc2-positive cells were unusually observed in the ulcerative region. CONCLUSIONS The Gαq/11-mediated pathway plays a pivotal role in the preservation of intestinal homeostasis, especially in Paneth cell maturation and positioning. Wnt/β-catenin signaling was reduced significantly in the crypt base in Gαq/G11-deficient mice, resulting in the defective maturation of Paneth cells, induction of differentiation toward goblet cells, and susceptibility to DSS colitis.
Collapse
Key Words
- Atoh1, atonal homolog 1
- BrdU, bromodeoxyuridine
- DSS, dextran sodium sulfate
- Defa1, defensin α1
- Dll1, delta-like 1
- FGF, fibroblast growth factor
- Fzd, frizzled
- Gna11
- Gnaq
- Hes, hairy/enhancer of split
- IEC, intestinal epithelial cell
- Ihh, Indian hedgehog
- Intermediate Cell
- NICD, Notch intracellular cytoplasmic domain
- PAS, periodic acid–Schiff
- PCR, polymerase chain reaction
- PKC, protein kinase C
- Paneth Cell
- TEM, transmission electron micrograph
- TUNEL, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling
- Tcf, T-cell factor
- Wnt
- mRNA, messenger RNA
- qPCR, quantitative real-time polymerase chain reaction
Collapse
|
21
|
Wang Y, Li Y, He Y, Sun Y, Sun W, Xie Q, Yin G, Du Y, Wang L, Shi G. Expression of G protein αq Subunit is Decreased in Lymphocytes from Patients with Rheumatoid Arthritis and is Correlated with Disease Activity. Scand J Immunol 2015; 75:203-9. [PMID: 21923740 DOI: 10.1111/j.1365-3083.2011.02635.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Gαq, the alpha subunit of Gq, a member of the Gq/11 sub-family, was reported to inhibit phosphatidylinositol-3-Kinase (PI3K) activation and prevent the activation of Akt. Previous studies demonstrated that mice losing Gαq in their immune system could spontaneously develop inflammatory arthritis. In this study, we showed that the Gαq expressions at mRNA and protein levels in the peripheral blood lymphocytes (PBLs) from patients with rheumatoid arthritis (RA) were significantly decreased in comparison of which in healthy individuals. The expression levels of Gαq mRNA in PBLs from patients with RA were correlated with RA disease activity (DAS28), anti-cyclic citrullinated protein antibodies, C-reactive protein and rheumatoid factor. We also demonstrated that Gαq controlled the apoptosis of RA PBLs through regulating the activity of Mcl-1 and caspase-3. These data suggested that Gαq might be involved in the pathogenesis of RA by regulating PBLs apoptosis.
Collapse
Affiliation(s)
- Y Wang
- Division of Rheumatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, ChinaGLP Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Y Li
- Division of Rheumatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, ChinaGLP Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Y He
- Division of Rheumatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, ChinaGLP Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Y Sun
- Division of Rheumatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, ChinaGLP Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - W Sun
- Division of Rheumatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, ChinaGLP Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Q Xie
- Division of Rheumatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, ChinaGLP Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - G Yin
- Division of Rheumatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, ChinaGLP Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Y Du
- Division of Rheumatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, ChinaGLP Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - L Wang
- Division of Rheumatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, ChinaGLP Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - G Shi
- Division of Rheumatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, ChinaGLP Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
GNA15 expression in small intestinal neuroendocrine neoplasia: Functional and signalling pathway analyses. Cell Signal 2015; 27:899-907. [DOI: 10.1016/j.cellsig.2015.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 01/18/2015] [Accepted: 02/02/2015] [Indexed: 11/23/2022]
|
23
|
Tsitoura P, Koussis K, Iatrou K. Inhibition of Anopheles gambiae odorant receptor function by mosquito repellents. J Biol Chem 2015; 290:7961-72. [PMID: 25657000 PMCID: PMC4367294 DOI: 10.1074/jbc.m114.632299] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 01/28/2015] [Indexed: 11/06/2022] Open
Abstract
The identification of molecular targets of insect repellents has been a challenging task, with their effects on odorant receptors (ORs) remaining a debatable issue. Here, we describe a study on the effects of selected mosquito repellents, including the widely used repellent N,N-diethyl-meta-toluamide (DEET), on the function of specific ORs of the African malaria vector Anopheles gambiae. This study, which has been based on quantitative measurements of a Ca(2+)-activated photoprotein biosensor of recombinant OR function in an insect cell-based expression platform and a sequential compound addition protocol, revealed that heteromeric OR (ORx/Orco) function was susceptible to strong inhibition by all tested mosquito repellents except DEET. Moreover, our results demonstrated that the observed inhibition was due to efficient blocking of Orco (olfactory receptor coreceptor) function. This mechanism of repellent action, which is reported for the first time, is distinct from the mode of action of other characterized insect repellents including DEET.
Collapse
Affiliation(s)
- Panagiota Tsitoura
- From the Insect Molecular Genetics and Biotechnology Group, Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", 15310 Athens and
| | - Konstantinos Koussis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 70013 Heraklion, Crete, Greece
| | - Kostas Iatrou
- From the Insect Molecular Genetics and Biotechnology Group, Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", 15310 Athens and
| |
Collapse
|
24
|
Hydroxyurea-inducible SAR1 gene acts through the Giα/JNK/Jun pathway to regulate γ-globin expression. Blood 2014; 124:1146-56. [PMID: 24914133 DOI: 10.1182/blood-2013-10-534842] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Hydroxyurea (HU) is effectively used in the management of β-hemoglobinopathies by augmenting the production of fetal hemoglobin (HbF). However, the molecular mechanisms underlying HU-mediated HbF regulation remain unclear. We previously reported that overexpression of the HU-induced SAR1 gene closely mimics the known effects of HU on K562 and CD34(+) cells, including γ-globin induction and cell-cycle regulation. Here, we show that HU stimulated nuclear factor-κB interaction with its cognate-binding site on the SAR1 promoter to regulate transcriptional expression of SAR1 in K562 and CD34(+) cells. Silencing SAR1 expression not only significantly lowered both basal and HU-elicited HbF production in K562 and CD34(+) cells, but also significantly reduced HU-mediated S-phase cell-cycle arrest and apoptosis in K562 cells. Inhibition of c-Jun N-terminal kinase (JNK)/Jun phosphorylation and silencing of Giα expression in SAR1-transfected K562 and CD34(+) cells reduced both γ-globin expression and HbF level, indicating that activation of Giα/JNK/Jun proteins is required for SAR1-mediated HbF induction. Furthermore, reciprocal coimmunoprecipitation assays revealed an association between forcibly expressed SAR1 and Giα2 or Giα3 proteins in both K562 and nonerythroid cells. These results indicate that HU induces SAR1, which in turn activates γ-globin expression, predominantly through the Giα/JNK/Jun pathway. Our findings identify SAR1 as an alternative therapeutic target for β-globin disorders.
Collapse
|
25
|
El-Haibi CP, Sharma P, Singh R, Gupta P, Taub DD, Singh S, Lillard JW. Differential G protein subunit expression by prostate cancer cells and their interaction with CXCR5. Mol Cancer 2013; 12:64. [PMID: 23773523 PMCID: PMC3720210 DOI: 10.1186/1476-4598-12-64] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 06/05/2013] [Indexed: 11/16/2022] Open
Abstract
Background Prostate cancer (PCa) cell lines and tissues differentially express CXCR5, which positively correlate with PCa progression, and mediate PCa cell migration and invasion following interaction with CXCL13. However, the differential expression of G protein α, β, and γ subunits by PCa cell lines and the precise combination of these proteins with CXCR5 has not been elucidated. Methods We examined differences in G protein expression of normal prostate (RWPE-1) and PCa cell lines (LNCaP, C4-2B, and PC3) by western blot analysis. Further, we immunoprecipitated CXCR5 with different G protein subunits, and CXCR4, following CXCL13 stimulation. To investigate constitutive coupling of CXCR5 with CXCR4 and PAR-1 we performed invasion assay in PCa cells transfected with Gαq/i2 or Gα13 siRNA, following CXCL13 treatment. We also investigated Rac and RhoA activity by G-LISA activation assay in PCa cells following CXCL13/thrombin stimulation. Result Of the 22 G proteins studied, Gαi1-3, Gβ1-4, Gγ5, Gγ7, and Gγ10 were expressed by both normal and PCa cell lines. Gαs was moderately expressed in C4-2B and PC3 cell lines, Gαq/11 was only present in RWPE-1 and LNCaP cell lines, while Gα12 and Gα13 were expressed in C4-2B and PC3 cell lines. Gγ9 was expressed only in PCa cell lines. Gα16, Gβ5, Gγ1-4, and Gγ13 were not detected in any of the cell lines studied. Surprisingly, CXCR4 co-immunoprecipitated with CXCR5 in PCa cell lines irrespective of CXCL13 treatment. We also identified specific G protein isoforms coupled to CXCR5 in its resting and active states. Gαq/11/Gβ3/Gγ9 in LNCaP and Gαi2/Gβ3/Gγ9 in C4-2B and PC3 cell lines, were coupled to CXCR5 and disassociated following CXCL13 stimulation. Interestingly, Gα13 co-immunoprecipitated with CXCR5 in CXCL13-treated, but not in untreated PCa cell lines. Inhibition of Gαq/i2 significantly decreased the ability of cells to invade, whereas silencing Gα13 did not affect CXCL13-dependent cell invasion. Finally, CXCL13 treatment significantly increased Rac activity in Gαq/i2 dependent manner, but not RhoA activity, in PCa cell lines. Conclusions These findings offer insight into molecular mechanisms of PCa progression and can help to design some therapeutic strategies involving CXCR5 and/or CXCL13 blockade and specific G protein inhibition to abrogate PCa metastasis.
Collapse
|
26
|
Čokić VP, Smith RD, Biancotto A, Noguchi CT, Puri RK, Schechter AN. Globin gene expression in correlation with G protein-related genes during erythroid differentiation. BMC Genomics 2013; 14:116. [PMID: 23425329 PMCID: PMC3602204 DOI: 10.1186/1471-2164-14-116] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 02/11/2013] [Indexed: 12/22/2022] Open
Abstract
Background The guanine nucleotide binding protein (G protein)-coupled receptors (GPCRs) regulate cell growth, proliferation and differentiation. G proteins are also implicated in erythroid differentiation, and some of them are expressed principally in hematopoietic cells. GPCRs-linked NO/cGMP and p38 MAPK signaling pathways already demonstrated potency for globin gene stimulation. By analyzing erythroid progenitors, derived from hematopoietic cells through in vitro ontogeny, our study intends to determine early markers and signaling pathways of globin gene regulation and their relation to GPCR expression. Results Human hematopoietic CD34+ progenitors are isolated from fetal liver (FL), cord blood (CB), adult bone marrow (BM), peripheral blood (PB) and G-CSF stimulated mobilized PB (mPB), and then differentiated in vitro into erythroid progenitors. We find that growth capacity is most abundant in FL- and CB-derived erythroid cells. The erythroid progenitor cells are sorted as 100% CD71+, but we did not find statistical significance in the variations of CD34, CD36 and GlyA antigens and that confirms similarity in maturation of studied ontogenic periods. During ontogeny, beta-globin gene expression reaches maximum levels in cells of adult blood origin (176 fmol/μg), while gamma-globin gene expression is consistently up-regulated in CB-derived cells (60 fmol/μg). During gamma-globin induction by hydroxycarbamide, we identify stimulated GPCRs (PTGDR, PTGER1) and GPCRs-coupled genes known to be activated via the cAMP/PKA (ADIPOQ), MAPK pathway (JUN) and NO/cGMP (PRPF18) signaling pathways. During ontogeny, GPR45 and ARRDC1 genes have the most prominent expression in FL-derived erythroid progenitor cells, GNL3 and GRP65 genes in CB-derived cells (high gamma-globin gene expression), GPR110 and GNG10 in BM-derived cells, GPR89C and GPR172A in PB-derived cells, and GPR44 and GNAQ genes in mPB-derived cells (high beta-globin gene expression). Conclusions These results demonstrate the concomitant activity of GPCR-coupled genes and related signaling pathways during erythropoietic stimulation of globin genes. In accordance with previous reports, the stimulation of GPCRs supports the postulated connection between cAMP/PKA and NO/cGMP pathways in activation of γ-globin expression, via JUN and p38 MAPK signaling.
Collapse
Affiliation(s)
- Vladan P Čokić
- Laboratory of Experimental Hematology, Institute for Medical Research, University of Belgrade, Dr, Subotica 4, 11129, Belgrade, Serbia.
| | | | | | | | | | | |
Collapse
|
27
|
Adjobo-Hermans MJ, Crosby KC, Putyrski M, Bhageloe A, van Weeren L, Schultz C, Goedhart J, Gadella TW. PLCβ isoforms differ in their subcellular location and their CT-domain dependent interaction with Gαq. Cell Signal 2013; 25:255-63. [DOI: 10.1016/j.cellsig.2012.09.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 09/08/2012] [Accepted: 09/16/2012] [Indexed: 11/15/2022]
|
28
|
Schepetkin IA, Kirpotina LN, Khlebnikov AI, Leopoldo M, Lucente E, Lacivita E, De Giorgio P, Quinn MT. 3-(1H-indol-3-yl)-2-[3-(4-nitrophenyl)ureido]propanamide enantiomers with human formyl-peptide receptor agonist activity: molecular modeling of chiral recognition by FPR2. Biochem Pharmacol 2012; 85:404-16. [PMID: 23219934 DOI: 10.1016/j.bcp.2012.11.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 11/14/2012] [Accepted: 11/15/2012] [Indexed: 01/14/2023]
Abstract
N-formyl peptide receptors (FPRs) are G protein-coupled receptors (GPCRs) that play critical roles in inflammatory reactions, and FPR-specific interactions can possibly be used to facilitate the resolution of pathological inflammatory reactions. Recent studies indicated that FPRs have stereo-selective preference for chiral ligands. Here, we investigated the structure-activity relationship of 24 chiral ureidopropanamides, including previously reported compounds PD168368/PD176252 and their close analogs, and used molecular modeling to define chiral recognition by FPR2. Unlike previously reported 6-methyl-2,4-disubstituted pyridazin-3(2H)-ones, whose R-forms preferentially activated FPR1/FPR2, we found that four S-enantiomers in the seven ureidopropanamide pairs tested preferentially activated intracellular Ca(2+) flux in FPR2-transfected cells, while the R-counterpart was more active in two enantiomer pairs. Thus, active enantiomers of FPR2 agonists can be in either R- or S-configurations, depending on the molecular scaffold and specific substituents at the chiral center. Using molecular modeling approaches, including field point methodology, homology modeling, and docking studies, we propose a model that can explain stereoselective activity of chiral FPR2 agonists. Importantly, our docking studies of FPR2 chiral agonists correlated well with the FPR2 pharmacophore model derived previously. We conclude that the ability of FPR2 to discriminate between the enantiomers is the consequence of the arrangement of the three asymmetric hydrophobic subpockets at the main orthosteric FPR2 binding site with specific orientation of charged regions in the subpockets.
Collapse
Affiliation(s)
- Igor A Schepetkin
- Department of Immunology and Infectious Diseases, Montana State University, Bozeman, MT 59717, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Lee MMK, Chui RKS, Tam IYS, Lau AHY, Wong YH. CCR1-mediated STAT3 tyrosine phosphorylation and CXCL8 expression in THP-1 macrophage-like cells involve pertussis toxin-insensitive Gα(14/16) signaling and IL-6 release. THE JOURNAL OF IMMUNOLOGY 2012; 189:5266-76. [PMID: 23125416 DOI: 10.4049/jimmunol.1103359] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Agonists of CCR1 contribute to hypersensitivity reactions and atherosclerotic lesions, possibly via the regulation of the transcription factor STAT3. CCR1 was demonstrated to use pertussis toxin-insensitive Gα(14/16) to stimulate phospholipase Cβ and NF-κB, whereas both Gα(14) and Gα(16) are also capable of activating STAT3. The coexpression of CCR1 and Gα(14/16) in human THP-1 macrophage-like cells suggests that CCR1 may use Gα(14/16) to induce STAT3 activation. In this study, we demonstrated that a CCR1 agonist, leukotactin-1 (CCL15), could indeed stimulate STAT3 Tyr(705) and Ser(727) phosphorylation via pertussis toxin-insensitive G proteins in PMA-differentiated THP-1 cells, human erythroleukemia cells, and HEK293 cells overexpressing CCR1 and Gα(14/16). The STAT3 Tyr(705) and Ser(727) phosphorylations were independent of each other and temporally distinct. Subcellular fractionation and confocal microscopy illustrated that Tyr(705)-phosphorylated STAT3 translocated to the nucleus, whereas Ser(727)-phosphorylated STAT3 was retained in the cytosol after CCR1/Gα(14) activation. CCL15 was capable of inducing IL-6 and IL-8 (CXCL8) production in both THP-1 macrophage-like cells and HEK293 cells overexpressing CCR1 and Gα(14/16). Neutralizing Ab to IL-6 inhibited CCL15-mediated STAT3 Tyr(705) phosphorylation, whereas inhibition of STAT3 activity abolished CCL15-activated CXCL8 release. The ability of CCR1 to signal through Gα(14/16) provides a linkage for CCL15 to regulate IL-6/STAT3-signaling cascades, leading to expression of CXCL8, a cytokine that is involved in inflammation and the rupture of atherosclerotic plaque.
Collapse
Affiliation(s)
- Maggie M K Lee
- Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | | | | | | | | |
Collapse
|
30
|
Choi SG, Jia J, Pfeffer RL, Sealfon SC. G proteins and autocrine signaling differentially regulate gonadotropin subunit expression in pituitary gonadotrope. J Biol Chem 2012; 287:21550-60. [PMID: 22549790 DOI: 10.1074/jbc.m112.348607] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Gonadotropin-releasing hormone (GnRH) acts at gonadotropes to direct the synthesis of the gonadotropins, follicle-stimulating hormone (FSH), and luteinizing hormone (LH). The frequency of GnRH pulses determines the pattern of gonadotropin synthesis. Several hypotheses for how the gonadotrope decodes GnRH frequency to regulate gonadotropin subunit genes differentially have been proposed. However, key regulators and underlying mechanisms remain uncertain. We investigated the role of individual G proteins by perturbations using siRNA or bacterial toxins. In LβT2 gonadotrope cells, FSHβ gene induction depended predominantly on Gα(q/11), whereas LHβ expression depended on Gα(s). Specifically reducing Gα(s) signaling also disinhibited FSHβ expression, suggesting the presence of a Gα(s)-dependent signal that suppressed FSH biosynthesis. The presence of secreted factors influencing FSHβ expression levels was tested by studying the effects of conditioned media from Gα(s) knockdown and cholera toxin-treated cells on FSHβ expression. These studies and related Transwell culture experiments implicate Gα(s)-dependent secreted factors in regulating both FSHβ and LHβ gene expression. siRNA studies identify inhibinα as a Gα(s)-dependent GnRH-induced autocrine regulatory factor that contributes to feedback suppression of FSHβ expression. These results uncover differential regulation of the gonadotropin genes by Gα(q/11) and by Gα(s) and implicate autocrine and gonadotrope-gonadotrope paracrine regulatory loops in the differential induction of gonadotropin genes.
Collapse
Affiliation(s)
- Soon-Gang Choi
- Center for Translational Systems Biology and the Department of Neurology, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | |
Collapse
|
31
|
|
32
|
Deng Y, Zhang W, Farhat K, Oberland S, Gisselmann G, Neuhaus EM. The stimulatory Gα(s) protein is involved in olfactory signal transduction in Drosophila. PLoS One 2011; 6:e18605. [PMID: 21490930 PMCID: PMC3072409 DOI: 10.1371/journal.pone.0018605] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Accepted: 03/11/2011] [Indexed: 11/19/2022] Open
Abstract
Seven-transmembrane receptors typically mediate olfactory signal transduction by coupling to G-proteins. Although insect odorant receptors have seven transmembrane domains like G-protein coupled receptors, they have an inverted membrane topology, constituting a key difference between the olfactory systems of insects and other animals. While heteromeric insect ORs form ligand-activated non-selective cation channels in recombinant expression systems, the evidence for an involvement of cyclic nucleotides and G-proteins in odor reception is inconsistent. We addressed this question in vivo by analyzing the role of G-proteins in olfactory signaling using electrophysiological recordings. We found that Gα(s) plays a crucial role for odorant induced signal transduction in OR83b expressing olfactory sensory neurons, but not in neurons expressing CO₂ responsive proteins GR21a/GR63a. Moreover, signaling of Drosophila ORs involved Gα(s) also in a heterologous expression system. In agreement with these observations was the finding that elevated levels of cAMP result in increased firing rates, demonstrating the existence of a cAMP dependent excitatory signaling pathway in the sensory neurons. Together, we provide evidence that Gα(s) plays a role in the OR mediated signaling cascade in Drosophila.
Collapse
Affiliation(s)
- Ying Deng
- Sino-France Joint Center for Drug Research and Screening, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, P. R. China
- Cell Physiology, Ruhr-University Bochum, Bochum, Germany
| | - Weiyi Zhang
- Cell Physiology, Ruhr-University Bochum, Bochum, Germany
- Bioduro (Beijing) Co. Ltd, Zhongguancun Life Science Park, Changping, Beijing, China
| | - Katja Farhat
- Cell Physiology, Ruhr-University Bochum, Bochum, Germany
- Department of Cardiovascular Physiology, Georg-August University, Göttingen, Germany
| | | | | | | |
Collapse
|
33
|
|
34
|
Galpha16 activates Ras by forming a complex with tetratricopeptide repeat 1 (TPR1) and Son of Sevenless (SOS). Cell Signal 2010; 22:1448-58. [PMID: 20639119 DOI: 10.1016/j.cellsig.2010.05.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Accepted: 05/23/2010] [Indexed: 11/24/2022]
Abstract
Many G protein-coupled receptors (GPCRs) are known to modulate cell growth and differentiation by stimulating the extracellular signal-regulated protein kinases (ERKs). In growth factor signaling, ERKs are typically stimulated through an elaborate network of modules consisting of adaptors, protein kinases, and the small GTPase Ras. The mechanism by which G protein signals tap into the ERK signaling pathway has thus far remain elusive. Members of the Gq family of G proteins, in particular Galpha16, have been shown to associate with tetratricopeptide repeat 1 (TPR1), an adaptor protein which preferentially binds to Ras. Here, we examined if TPR1 is indeed the missing link between Galpha16 signaling and Ras activation. Expression of Galpha16QL, a constitutively active mutant of Galpha16, in HEK 293 cells led to the formation of GTP-bound Ras and the subsequent phosphorylation of ERK. Likewise, stimulation of endogenou G16-coupled CCR1 chemokine receptors produced the same responses in human erythroleukemia cells. siRNA-mediated knockdown of TPR1 or expression of a dominant negative mutant of TPR1 effectively abolished the ability of Galpha16QL to induce Ras activation in HEK 293 cells. In contrast, these manipulations had no inhibitory effect on Galpha16QL induced activation of phospholipase Cbeta. Galpha16QL-induced phosphorylations of downstream targets including ERK, signal transducer and activator of transcription 3, and IkappaB kinase were significantly suppressed upon expression of the dominant negative mutant of TPR1. Furthermore, SOS2, a Ras guanine nucleotide exchange factor, was found to form a complex with TPR1 and Galpha16QL. Expression of SOS2 enhanced Galpha16QL-induced Ras activation and its subsequent signaling. Collectively, our results suggest that Galpha16 regulates multiple signaling pathways by activating Ras through its association with TPR1, but TPR1 is not required for Galpha16 to stimulate phospholipase Cbeta.
Collapse
|
35
|
Franco R, Canela EI, Casado V, Ferre S. Platforms for the identification of GPCR targets, and of orthosteric and allosteric modulators. Expert Opin Drug Discov 2010; 5:391-403. [DOI: 10.1517/17460441003653163] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
36
|
Kara FM, Chitu V, Sloane J, Axelrod M, Fredholm BB, Stanley ER, Cronstein BN. Adenosine A1 receptors (A1Rs) play a critical role in osteoclast formation and function. FASEB J 2010; 24:2325-33. [PMID: 20181934 DOI: 10.1096/fj.09-147447] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Adenosine regulates a wide variety of physiological processes via interaction with one or more G-protein-coupled receptors (A(1)R, A(2A)R, A(2B)R, and A(3)R). Because A(1)R occupancy promotes fusion of human monocytes to form giant cells in vitro, we determined whether A(1)R occupancy similarly promotes osteoclast function and formation. Bone marrow cells (BMCs) were harvested from C57Bl/6 female mice or A(1)R-knockout mice and their wild-type (WT) littermates and differentiated into osteoclasts in the presence of colony stimulating factor-1 and receptor activator of NF-kappaB ligand in the presence or absence of the A(1)R antagonist 1,3-dipropyl-8-cyclopentyl xanthine (DPCPX). Osteoclast morphology was analyzed in tartrate-resistant acid phosphatase or F-actin-stained samples, and bone resorption was evaluated by toluidine blue staining of dentin. BMCs from A(1)R-knockout mice form fewer osteoclasts than BMCs from WT mice, and the A(1)R antagonist DPCPX inhibits osteoclast formation (IC(50)=1 nM), with altered morphology and reduced ability to resorb bone. A(1)R blockade increased ubiquitination and degradation of TRAF6 in RAW264.7 cells induced to differentiate into osteoclasts. These studies suggest a critical role for adenosine in bone homeostasis via interaction with adenosine A(1)R and further suggest that A(1)R may be a novel pharmacologic target to prevent the bone loss associated with inflammatory diseases and menopause.
Collapse
Affiliation(s)
- Firas M Kara
- Department of Medicine, Division of Clinical Pharmacology, NYU School of Medicine, 550 First Ave., New York, NY 10016, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Lee MMK, Wong YH. CCR1-mediated activation of nuclear factor-κB in THP-1 monocytic cells involvespertussistoxin-insensitive Gα14and Gα16signaling cascades. J Leukoc Biol 2009; 86:1319-29. [DOI: 10.1189/jlb.0209052] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
38
|
Ye RD, Boulay F, Wang JM, Dahlgren C, Gerard C, Parmentier M, Serhan CN, Murphy PM. International Union of Basic and Clinical Pharmacology. LXXIII. Nomenclature for the formyl peptide receptor (FPR) family. Pharmacol Rev 2009; 61:119-61. [PMID: 19498085 PMCID: PMC2745437 DOI: 10.1124/pr.109.001578] [Citation(s) in RCA: 618] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Formyl peptide receptors (FPRs) are a small group of seven-transmembrane domain, G protein-coupled receptors that are expressed mainly by mammalian phagocytic leukocytes and are known to be important in host defense and inflammation. The three human FPRs (FPR1, FPR2/ALX, and FPR3) share significant sequence homology and are encoded by clustered genes. Collectively, these receptors bind an extraordinarily numerous and structurally diverse group of agonistic ligands, including N-formyl and nonformyl peptides of different composition, that chemoattract and activate phagocytes. N-formyl peptides, which are encoded in nature only by bacterial and mitochondrial genes and result from obligatory initiation of bacterial and mitochondrial protein synthesis with N-formylmethionine, is the only ligand class common to all three human receptors. Surprisingly, the endogenous anti-inflammatory peptide annexin 1 and its N-terminal fragments also bind human FPR1 and FPR2/ALX, and the anti-inflammatory eicosanoid lipoxin A4 is an agonist at FPR2/ALX. In comparison, fewer agonists have been identified for FPR3, the third member in this receptor family. Structural and functional studies of the FPRs have produced important information for understanding the general pharmacological principles governing all leukocyte chemoattractant receptors. This article aims to provide an overview of the discovery and pharmacological characterization of FPRs, to introduce an International Union of Basic and Clinical Pharmacology (IUPHAR)-recommended nomenclature, and to discuss unmet challenges, including the mechanisms used by these receptors to bind diverse ligands and mediate different biological functions.
Collapse
Affiliation(s)
- Richard D Ye
- Department of Pharmacology, University of Illinois College of Medicine, 835 South Wolcott Avenue, M/C 868, Chicago, Illinois 60612, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Tian Y, Lee MM, Yung LY, Allen RA, Slocombe PM, Twomey BM, Wong YH. Differential involvement of Gα16 in CC chemokine-induced stimulation of phospholipase Cβ, ERK, and chemotaxis. Cell Signal 2008; 20:1179-89. [DOI: 10.1016/j.cellsig.2008.02.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2007] [Revised: 02/05/2008] [Accepted: 02/13/2008] [Indexed: 01/27/2023]
|
40
|
Constitutive serum response factor activation by the viral chemokine receptor homologue pUS28 is differentially regulated by Galpha(q/11) and Galpha(16). Cell Signal 2008; 20:1528-37. [PMID: 18534820 DOI: 10.1016/j.cellsig.2008.04.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2008] [Accepted: 04/11/2008] [Indexed: 01/30/2023]
Abstract
Expression of the human cytomegalovirus (HCMV)-encoded chemokine receptor homologue pUS28 in mammalian cells results in ligand-dependent and -independent changes in the activity of multiple cellular signal transduction pathways. The ligand-dependent signalling activity of pUS28 has been shown to be predominantly mediated by heterotrimeric G proteins of the G(i/o) and G(12/13) subfamilies. Ligand-independent constitutive activity of pUS28 causing stimulation of inositol phosphate formation has been correlated with the coupling of pUS28 to G proteins of the G(q) family. It is well known that activation of G(q) proteins by cell surface receptors is coupled to activation of the Rho GTPase RhoA. Activated RhoA regulates numerous cellular functions, including the activity of the transcription factor serum response factor (SRF). The marked activation of G(q) proteins by pUS28 in transfected and HCMV-infected cells prompted us to investigate its effect on SRF activity. The results presented herein demonstrate that expression of pUS28 in COS-7 cells caused a vigorous induction of SRF activity. This effect was observed in the absence of chemokines known to interact with pUS28, and was specifically mediated by endogenous G(q) and/or G(11) as well as RhoA and/or a closely related Rho GTPase. The stimulatory effect of pUS28 and Galpha(q/11) was independent of phospholipase C-beta (PLCbeta) activation and was markedly sensitive to inhibition by wild-type, but not by constitutively active Galpha(16), thus identifying Galpha(16) as a modulator of Galpha(q/11) function likely to act by competing with Galpha(q/11) for and thus uncoupling Galpha(q/11) from activation by pUS28.
Collapse
|
41
|
Matsumoto Y, Funahashi J, Mori K, Hayashi K, Yano H. The noncompetitive antagonism of histamine H1 receptors expressed in Chinese hamster ovary cells by olopatadine hydrochloride: its potency and molecular mechanism. Pharmacology 2008; 81:266-74. [PMID: 18268402 DOI: 10.1159/000115970] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2007] [Accepted: 11/13/2007] [Indexed: 11/19/2022]
Abstract
Calcium responses to various concentrations of histamine were monitored in Chinese hamster ovary cells stably expressing the human histamine H(1) receptor. The effects of various histamine H(1) receptor antagonists on the dose-response curve for histamine were evaluated. Olopatadine hydrochloride (olopatadine) inhibited the histamine-induced maximum response (pD(2)': 7.5) but had insignificant effects on histamine EC(50) values. This noncompetitive property exhibited by olopatadine, which was also observed in human umbilical vein endothelial cells, was the most striking among the antihistamines tested in this study. The geometrical isomer of olopatadine (E-isomer), which had a similar binding affinity to the histamine H(1) receptor as olopatadine, showed a mixed antagonistic profile (competitive and noncompetitive). These results indicate that the geometry around the double bond in the dimethylaminopropylidene group is critical for the potent noncompetitive property of olopatadine. Furthermore, binding mode analyses suggest that the protonated amine group in the dimethylaminopropylidene moiety of olopatadine forms an ionic bond with Glu 181 that is present in the second extracellular loop of the histamine H(1) receptor, whereas the amine group of the E-isomer does not. The second extracellular loop in aminergic G-protein-coupled receptors contributes to ligand binding and therefore the noncompetitive property of olopatadine may be explained by the interaction with Glu 181.
Collapse
Affiliation(s)
- Yuichi Matsumoto
- Pharmaceutical Research Center, Kyowa Hakko Kogyo Co., Ltd., Japan
| | | | | | | | | |
Collapse
|
42
|
Bruges G, Borges A, Sánchez de Villarroel S, Lippo de Bécemberg I, Francis de Toba G, Pláceres F, González de Alfonzo R, Alfonzo MJ. Coupling of M3 acetylcholine receptor to Gq16 activates a natriuretic peptide receptor guanylyl cyclase. J Recept Signal Transduct Res 2007; 27:189-216. [PMID: 17613728 DOI: 10.1080/10799890701417899] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Muscarinic activation of tracheal smooth muscle (TSM) involves a M(3)AChR/heterotrimeric-G protein/NPR-GC coupling mechanism. G protein activators Mastoparan (MAS) and Mastoparan-7 stimulated 4- and 10-fold the NPR-GC respectively, being insensitive to PTX and antibodies against Galpha(i/o) subfamily. Muscarinic and MAS stimulation of NPR-GC was blocked by antibodies against C-terminal of Galpha(q16), whose expression was confirmed by RT-PCR. However, synthetic peptides from C-terminal of Galpha(q15/16) stimulated the NPR-GC. Coupling of alpha(q16) to M(3)AChR is supported by MAS decreased [(3)H]QNB binding, being abolished after M(3)AChR-4-DAMP-alkylation. Anti-i(3)M(3)AChR antibodies blocked the muscarinic activation of NPR-GC, and synthetic peptide from i(3)M(3)AChR (M(3)P) was more potent than MAS increasing GTPgamma [(35)S] and decreasing the [(3)H]QNB activities. Coupling between NPR-GC and Galpha(q16) was evaluated by using trypsin-solubilized-fraction from TSM membranes, which displayed a MAS-sensitive-NPR-GC activity, being immunoprecipitated with anti-Galpha(q16), also showing an immunoreactive heterotrimeric-G-beta-subunit. These data support the existence of a novel transducing cascade, involving Galpha(q16)beta gamma coupling M(3)AChR to NPR-GC.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies/pharmacology
- Blotting, Western
- Cattle
- Chromatography, Affinity
- Cytoplasm/drug effects
- Cytoplasm/metabolism
- Enzyme Activation/drug effects
- GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors
- GTP-Binding Protein alpha Subunits, Gq-G11/metabolism
- Guanosine Triphosphate/pharmacology
- Guanylate Cyclase/isolation & purification
- Guanylate Cyclase/metabolism
- Heterotrimeric GTP-Binding Proteins/antagonists & inhibitors
- Heterotrimeric GTP-Binding Proteins/metabolism
- Intercellular Signaling Peptides and Proteins
- Molecular Sequence Data
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Peptides/chemistry
- Peptides/pharmacology
- Protein Processing, Post-Translational/drug effects
- Receptor, Muscarinic M2/metabolism
- Receptor, Muscarinic M3/antagonists & inhibitors
- Receptor, Muscarinic M3/metabolism
- Receptors, Atrial Natriuretic Factor/metabolism
- Solubility/drug effects
- Trypsin/metabolism
- Wasp Venoms/chemistry
- Wasp Venoms/pharmacology
Collapse
Affiliation(s)
- Gustavo Bruges
- Sección de Biomembranas, Instituto de Medicina Experimental, Cátedra de Patología General y Fisiopatología, Escuela Luis Razetti, Facultad de Medicina, Universidad Central de Venezuela, Sabana Grande, Caracas, Venezuela
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Suga H, Haga T. Ligand screening system using fusion proteins of G protein-coupled receptors with G protein alpha subunits. Neurochem Int 2007; 51:140-64. [PMID: 17659814 DOI: 10.1016/j.neuint.2007.06.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2007] [Revised: 06/07/2007] [Accepted: 06/08/2007] [Indexed: 01/04/2023]
Abstract
G protein-coupled receptors (GPCRs) constitute one of the largest families of genes in the human genome, and are the largest targets for drug development. Although a large number of GPCR genes have recently been identified, ligands have not yet been identified for many of them. Various assay systems have been employed to identify ligands for orphan GPCRs, but there is still no simple and general method to screen for ligands of such GPCRs, particularly of G(i)-coupled receptors. We have examined whether fusion proteins of GPCRs with G protein alpha subunit (Galpha) could be utilized for ligand screening and showed that the fusion proteins provide an effective method for the purpose. This article focuses on the followings: (1) characterization of GPCR genes and GPCRs, (2) identification of ligands for orphan GPCRs, (3) characterization of GPCR-Galpha fusion proteins, and (4) identification of ligands for orphan GPCRs using GPCR-Galpha fusion proteins.
Collapse
Affiliation(s)
- Hinako Suga
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | | |
Collapse
|
44
|
Wang J, Wu X, Simonavicius N, Tian H, Ling L. Medium-chain fatty acids as ligands for orphan G protein-coupled receptor GPR84. J Biol Chem 2006; 281:34457-64. [PMID: 16966319 DOI: 10.1074/jbc.m608019200] [Citation(s) in RCA: 311] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Free fatty acids (FFAs) play important physiological roles in many tissues as an energy source and as signaling molecules in various cellular processes. Elevated levels of circulating FFAs are associated with obesity, dyslipidemia, and diabetes. Here we show that GPR84, a previously orphan G protein-coupled receptor, functions as a receptor for medium-chain FFAs with carbon chain lengths of 9-14. Medium-chain FFAs elicit calcium mobilization, inhibit 3',5'-cyclic AMP production, and stimulate [35S]guanosine 5'-O-(3-thiotriphosphate) binding in a GPR84-dependent manner. The activation of GPR84 by medium-chain FFAs couples primarily to a pertussis toxin-sensitive G(i/o) pathway. In addition, we show that GPR84 is selectively expressed in leukocytes and markedly induced in monocytes/macrophages upon activation by lipopolysaccharide. Furthermore, we demonstrate that medium-chain FFAs amplify lipopolysaccharide-stimulated production of the proinflammatory cytokine interleukin-12 p40 through GPR84. Our results indicate a role for GPR84 in directly linking fatty acid metabolism to immunological regulation.
Collapse
Affiliation(s)
- Jinghong Wang
- Amgen Inc., South San Francisco, California 94080, USA
| | | | | | | | | |
Collapse
|
45
|
Innamorati G, Piccirillo R, Bagnato P, Palmisano I, Schiaffino MV. The melanosomal/lysosomal protein OA1 has properties of a G protein-coupled receptor. ACTA ACUST UNITED AC 2006; 19:125-35. [PMID: 16524428 PMCID: PMC1459912 DOI: 10.1111/j.1600-0749.2006.00292.x] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The protein product of the ocular albinism type 1 gene, named OA1, is a pigment cell-specific integral membrane glycoprotein, localized to melanosomes and lysosomes and possibly implicated in melanosome biogenesis. Although its function remains unknown, we previously showed that OA1 shares structural similarities with G protein-coupled receptors (GPCRs). To ascertain the molecular function of OA1 and in particular its nature as a GPCR, we adopted a heterologous expression strategy commonly exploited to demonstrate GPCR-mediated signaling in mammalian cells. Here we show that when expressed in COS7 cells OA1 displays a considerable and spontaneous capacity to activate heterotrimeric G proteins and the associated signaling cascade. In contrast, OA1 mutants carrying either a missense mutation or a small deletion in the third cytosolic loop lack this ability. Furthermore, OA1 is phosphorylated and interacts with arrestins, well-established multifunctional adaptors of conformationally active GPCRs. In fact, OA1 colocalizes and coprecipitates with arrestins, which downregulate the signaling of OA1 by specifically reducing its expression levels. These findings indicate that heterologously expressed OA1 exhibits two fundamental properties of GPCRs, being capable to activate heterotrimeric G proteins and to functionally associate with arrestins, and provide proof of principle that OA1 can actually function as a canonical GPCR in mammalian cells.
Collapse
Affiliation(s)
- Giulio Innamorati
- DIBIT, Scientific Institute San Raffalele, Via Olgettina 58, 20132 Milan, Italy
| | | | | | | | | |
Collapse
|
46
|
Wang J, Simonavicius N, Wu X, Swaminath G, Reagan J, Tian H, Ling L. Kynurenic acid as a ligand for orphan G protein-coupled receptor GPR35. J Biol Chem 2006; 281:22021-22028. [PMID: 16754668 DOI: 10.1074/jbc.m603503200] [Citation(s) in RCA: 502] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Local catabolism of the essential amino acid tryptophan is considered an important mechanism in regulating immunological and neurological responses. The kynurenine pathway is the main route for the non-protein metabolism of tryptophan. The intermediates of the kynurenine pathway are present at micromolar concentrations in blood and are regulated by inflammatory stimuli. Here we show that GPR35, a previously orphan G protein-coupled receptor, functions as a receptor for the kynurenine pathway intermediate kynurenic acid. Kynurenic acid elicits calcium mobilization and inositol phosphate production in a GPR35-dependent manner in the presence of G(qi/o) chimeric G proteins. Kynurenic acid stimulates [35S]guanosine 5'-O-(3-thiotriphosphate) binding in GPR35-expressing cells, an effect abolished by pertussis toxin treatment. Kynurenic acid also induces the internalization of GPR35. Expression analysis indicates that GPR35 is predominantly detected in immune cells and the gastrointestinal tract. Furthermore, we show that kynurenic acid inhibits lipopolysaccharide-induced tumor necrosis factor-alpha secretion in peripheral blood mononuclear cells. Our results suggest unexpected signaling functions for kynurenic acid through GPR35 activation.
Collapse
Affiliation(s)
| | | | - Xiaosu Wu
- Amgen Inc., South San Francisco, California 94080
| | | | - Jeff Reagan
- Amgen Inc., South San Francisco, California 94080
| | - Hui Tian
- Amgen Inc., South San Francisco, California 94080
| | - Lei Ling
- Amgen Inc., South San Francisco, California 94080.
| |
Collapse
|
47
|
Lo RKH, Wong YH. Transcriptional activation of c-Fos by constitutively active Galpha(16)QL through a STAT1-dependent pathway. Cell Signal 2006; 18:2143-53. [PMID: 16781847 DOI: 10.1016/j.cellsig.2006.04.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2006] [Accepted: 04/25/2006] [Indexed: 02/06/2023]
Abstract
Hematopoietic restrictive Galpha(16) has long been known to stimulate phospholipase Cbeta (PLCbeta) and induce mitogen-activated protein kinase (MAPK) phosphorylation. Recently, we have demonstrated that Galpha(16) is capable of inducing the phosphorylation and transcriptional activation of transcription factors, such as signal transducer and activator of transcription 3 (STAT3) and nuclear factor kappaB (NFkappaB). However, the downstream signaling regulation by Galpha(16) has not yet been documented. In the present study, we have determined the signaling mechanism by which constitutively active Galpha(16) mediates c-Fos transcriptional activation in human embryonic kidney (HEK) 293 cells. Overexpression of constitutively active Galpha(16), Galpha(16)QL, resulted in the stimulation of c-Fos transcriptional activation in HEK 293 cells. The participation of PLCbeta, c-Src/Janus kinase 2 (JAK2) and extracellular signal-regulated kinase (ERK) signaling pathways in Galpha(16)QL-induced c-Fos transcriptional activation was demonstrated by the use of their specific inhibitors. However, c-Jun N terminal kinase (JNK), p38 MAPK and phosphatidylinositol-3 kinase (PI3K) were not required. Interestingly, the dominant negative mutant of STAT1, but not STAT3, suppressed c-Fos transcriptional activation induced by Galpha(16)QL, implying that STAT1 was involved in this signaling mechanism. To further examine the role of STAT1 in the signaling pathway of Galpha(16), we demonstrated that Galpha(16)QL was able to induce STAT1 activation. Also, stimulation of adenosine A1 receptor-coupled Galpha(16) was shown to induce ERK and STAT1 phosphorylations in a concentration-dependent manner. Using selective inhibitors, PLCbeta, c-Src/JAK and ERK, but not JNK, p38 MAPK and PI3K, were shown to be involved in Galpha(16)QL-induced STAT1 activation. Collectively, our results demonstrate for the first time that stimulation of Galpha(16) can lead to STAT1-dependent c-Fos transcriptional activation via PLCbeta, c-Src/JAK and ERK pathways.
Collapse
Affiliation(s)
- Rico K H Lo
- Department of Biochemistry, Molecular Neuroscience Center, and Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | | |
Collapse
|
48
|
Kucukkaya B, Arslan DO, Kan B. Role of G proteins and ERK activation in hemin-induced erythroid differentiation of K562 cells. Life Sci 2006; 78:1217-24. [PMID: 16216279 DOI: 10.1016/j.lfs.2005.06.041] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2005] [Accepted: 06/22/2005] [Indexed: 10/25/2022]
Abstract
Heterotrimeric G proteins which couple extracellular signals to intracellular effectors play a central role in cell growth and differentiation. The pluripotent erythroleukemic cell line K562 that acquires the capability to synthesize hemoglobin in response to a variety of agents can be used as a model system for erythroid differentiation. Using Western blot analysis and RT-PCR, we studied alterations in G protein expression accompanying hemin-induced differentiation of K562 cells. We demonstrated the presence of G(alpha s), G(alpha i2) and G(alpha q) and the absence of G(alpha i1), G(alpha o) and G(alpha 16) in K562 cells. We observed the short form of G(alpha s) to be expressed predominantly in these cells. Treatment of K562 cells with hemin resulted in an increase in the levels of G(alpha s) and G(alpha q). On the other hand, the level of G(alpha i2) was found to increase on the third day after induction with hemin, followed by a decrease to levels lower of those of uninduced cells. The mitogen-activated protein kinase ERK1/2 pathway is crucial in the control of cell proliferation and differentiation. Both Gi- and Gq-coupled receptors stimulate MAPK activation. We therefore examined the phosphorylation of ERK1/2 during hemin-induced differentiation of K562 cells. Using anti-ERK1/2 antibodies, we observed that ERK2 was primarily phosphorylated in K562 cells. ERK2 phosphorylation increased gradually until 48 h and returned to basal values by 96 h following hemin treatment. Our results suggest that changes in G protein expression and ERK2 activity are involved in hemin-induced differentiation of K562 cells.
Collapse
Affiliation(s)
- Bahire Kucukkaya
- Marmara University School of Medicine, Department of Biophysics, Tibbiye Caddesi No 49, Haydarpasa, 34668, Istanbul, Turkey
| | | | | |
Collapse
|
49
|
Johansson BB, Minsaas L, Aragay AM. Proteasome involvement in the degradation of the G(q) family of Galpha subunits. FEBS J 2005; 272:5365-77. [PMID: 16218966 DOI: 10.1111/j.1742-4658.2005.04934.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Metabolically unstable proteins are involved in a multitude of regulatory networks, including those that control cell signaling, the cell cycle and in many responses to physiological stress. In the present study, we have determined the stability and characterized the degradation process of some members of the G(q) class of heterotrimeric G proteins. Pulse-chase experiments in HEK293 cells indicated a rapid turnover of endogenously expressed Galpha(q) and overexpressed Galpha(q) and Galpha(16) subunits. Pretreatment with proteasome inhibitors attenuated the degradation of both G alpha subunits. In contrast, pretreatment of cells with inhibitors of lysosomal proteases and nonproteasomal cysteine proteases had very little effect on the stability of the proteins. Significantly, the turnover of these proteins is not affected by transient activation of their associated receptors. Fractionation studies showed that the rates of Galpha(q) and Galpha16 degradation are accelerated in the cytosol. In fact, we show that a mutant Galpha(q) which lacks its palmitoyl modification site, and which is localized almost entirely in the cytoplasm, has a marked increase in the rate of degradation. Taken together, these results suggest that the G(q) class proteins are degraded through the proteasome pathway and that cellular localization and/or other protein interactions determine their stability.
Collapse
Affiliation(s)
- Bente B Johansson
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Norway
| | | | | |
Collapse
|
50
|
Leifert WR, Aloia AL, Bucco O, Glatz RV, McMurchie EJ. G-protein-coupled receptors in drug discovery: nanosizing using cell-free technologies and molecular biology approaches. ACTA ACUST UNITED AC 2005; 10:765-79. [PMID: 16234342 DOI: 10.1177/1087057105280517] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Signal transduction by G-protein-coupled receptors (GPCRs) underpins a multitude of physiological processes. Ligand recognition by the receptor leads to activation of a generic molecular switch involving heterotrimeric G-proteins and guanine nucleotides. Signal transduction has been studied extensively with both cell-based systems and assays comprising isolated signaling components. Interest and commercial investment in GPCRs in areas such as drug targets, orphan receptors, high throughput screening, biosensors, and so on will focus greater attention on assay development to allow for miniaturization, ultra-high throughput and, eventually, microarray/biochip assay formats. Although cell-based assays are adequate for many GPCRs, it is likely that these formats will limit the development of higher density GPCR assay platforms mandatory for other applications. Stable, robust, cell-free signaling assemblies comprising receptor and appropriate molecular switching components will form the basis of future GPCR assay platforms adaptable for such applications as microarrays. The authors review current cell-free GPCR assay technologies and molecular biological approaches for construction of novel, functional GPCR assays.
Collapse
Affiliation(s)
- Wayne R Leifert
- CSIRO Molecular and Health Technologies, Adelaide, SA, Australia.
| | | | | | | | | |
Collapse
|