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
|
Todd TD, Vithani N, Singh S, Bowman GR, Blumer KJ, Soranno A. Stabilization of interdomain closure by a G protein inhibitor. Proc Natl Acad Sci U S A 2024; 121:e2311711121. [PMID: 39196624 PMCID: PMC11388362 DOI: 10.1073/pnas.2311711121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 05/29/2024] [Indexed: 08/29/2024] Open
Abstract
Inhibitors of heterotrimeric G proteins are being developed as therapeutic agents. Epitomizing this approach are YM-254890 (YM) and FR900359 (FR), which are efficacious in models of thrombosis, hypertension, obesity, asthma, uveal melanoma, and pain, and under investigation as an FR-antibody conjugate in uveal melanoma clinical trials. YM/FR inhibits the Gq/11/14 subfamily by interfering with GDP (guanosine diphosphate) release, but by an unknown biophysical mechanism. Here, we show that YM inhibits GDP release by stabilizing closure between the Ras-like and α-helical domains of a Gα subunit. Nucleotide-free Gα adopts an ensemble of open and closed configurations, as indicated by single-molecule Förster resonance energy transfer and molecular dynamics simulations, whereas GDP and GTPγS (guanosine 5'-O-[gamma-thio]triphosphate) stabilize distinct closed configurations. YM stabilizes closure in the presence or absence of GDP without requiring an intact interdomain interface. All three classes of mammalian Gα subunits that are insensitive to YM/FR possess homologous but degenerate YM/FR binding sites, yet can be inhibited upon transplantation of the YM/FR binding site of Gq. Novel YM/FR analogs tailored to each class of G protein will provide powerful new tools for therapeutic investigation.
Collapse
Affiliation(s)
- Tyson D Todd
- Department of Cell Biology and Physiology, Washington University in St. Louis, Saint Louis, MO 63110
| | - Neha Vithani
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, Saint Louis, MO 63110
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104-6059
| | - Sukrit Singh
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, Saint Louis, MO 63110
| | - Gregory R Bowman
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, Saint Louis, MO 63110
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104-6059
| | - Kendall J Blumer
- Department of Cell Biology and Physiology, Washington University in St. Louis, Saint Louis, MO 63110
| | - Andrea Soranno
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, Saint Louis, MO 63110
- Department of Biochemistry and Biophysics, Center for Biomolecular Condensates, Washington University in St. Louis, Saint Louis, MO 63130
| |
Collapse
|
3
|
Bonifer C, Hanke W, Mühle J, Löhr F, Becker-Baldus J, Nagel J, Schertler GFX, Müller CE, König GM, Hilger D, Glaubitz C. Structural response of G protein binding to the cyclodepsipeptide inhibitor FR900359 probed by NMR spectroscopy. Chem Sci 2024; 15:12939-12956. [PMID: 39148790 PMCID: PMC11323312 DOI: 10.1039/d4sc01950d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 06/27/2024] [Indexed: 08/17/2024] Open
Abstract
The cyclodepsipeptide FR900359 (FR) and its analogs are able to selectively inhibit the class of Gq proteins by blocking GDP/GTP exchange. The inhibitor binding site of Gq has been characterized by X-ray crystallography, and various binding and functional studies have determined binding kinetics and mode of inhibition. Here we investigate isotope-labeled FR bound to the membrane-anchored G protein heterotrimer by solid-state nuclear magnetic resonance (ssNMR) and in solution by liquid-state NMR. The resulting data allowed us to identify regions of the inhibitor which show especially pronounced effects upon binding and revealed a generally rigid binding mode in the cis conformation under native-like conditions. The inclusion of the membrane environment allowed us to show a deep penetration of FR into the lipid bilayer illustrating a possible access mode of FR into the cell. Dynamic nuclear polarization (DNP)-enhanced ssNMR was used to observe the structural response of specific segments of the Gα subunit to inhibitor binding. This revealed rigidification of the switch I binding site and an allosteric response in the α5 helix as well as suppression of structural changes induced by nucleotide exchange due to inhibition by FR. Our NMR studies of the FR-G protein complex conducted directly within a native membrane environment provide important insights into the inhibitors access via the lipid membrane, binding mode, and structural allosteric effects.
Collapse
Affiliation(s)
- Christian Bonifer
- Institute of Biophysical Chemistry, Centre of Biomolecular Magnetic Resonance, Goethe University Frankfurt Max-von-Laue Str. 9 60438 Frankfurt Germany
| | - Wiebke Hanke
- Institute for Pharmaceutical Biology, University of Bonn Nussallee 6 53115 Bonn Germany
| | - Jonas Mühle
- Division of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institute Forschungsstr. 111, 5232 Villigen PSI Switzerland
| | - Frank Löhr
- Institute of Biophysical Chemistry, Centre of Biomolecular Magnetic Resonance, Goethe University Frankfurt Max-von-Laue Str. 9 60438 Frankfurt Germany
| | - Johanna Becker-Baldus
- Institute of Biophysical Chemistry, Centre of Biomolecular Magnetic Resonance, Goethe University Frankfurt Max-von-Laue Str. 9 60438 Frankfurt Germany
| | - Jessica Nagel
- Department of Pharmaceutical & Medicinal Chemistry, Pharmaceutical Institute, University of Bonn An der Immenburg 4 53121 Bonn Germany
| | - Gebhard F X Schertler
- Division of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institute Forschungsstr. 111, 5232 Villigen PSI Switzerland
| | - Christa E Müller
- Department of Pharmaceutical & Medicinal Chemistry, Pharmaceutical Institute, University of Bonn An der Immenburg 4 53121 Bonn Germany
| | - Gabriele M König
- Institute for Pharmaceutical Biology, University of Bonn Nussallee 6 53115 Bonn Germany
| | - Daniel Hilger
- Department of Pharmaceutical Chemistry, University of Marburg 35037 Marburg Germany
| | - Clemens Glaubitz
- Institute of Biophysical Chemistry, Centre of Biomolecular Magnetic Resonance, Goethe University Frankfurt Max-von-Laue Str. 9 60438 Frankfurt Germany
| |
Collapse
|
4
|
Ren J, Cui Z, Jiang C, Wang L, Guan Y, Ren Y, Zhang S, Tu T, Yu J, Li Y, Duan W, Guan J, Wang K, Zhang H, Xing D, Kahn ML, Zhang H, Hong T. GNA14 and GNAQ somatic mutations cause spinal and intracranial extra-axial cavernous hemangiomas. Am J Hum Genet 2024; 111:1370-1382. [PMID: 38917801 PMCID: PMC11267519 DOI: 10.1016/j.ajhg.2024.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024] Open
Abstract
Extra-axial cavernous hemangiomas (ECHs) are complex vascular lesions mainly found in the spine and cavernous sinus. Their removal poses significant risk due to their vascularity and diffuse nature, and their genetic underpinnings remain incompletely understood. Our approach involved genetic analyses on 31 tissue samples of ECHs employing whole-exome sequencing and targeted deep sequencing. We explored downstream signaling pathways, gene expression changes, and resultant phenotypic shifts induced by these mutations, both in vitro and in vivo. In our cohort, 77.4% of samples had somatic missense variants in GNA14, GNAQ, or GJA4. Transcriptomic analysis highlighted significant pathway upregulation, with the GNAQ c.626A>G (p.Gln209Arg) mutation elevating PI3K-AKT-mTOR and angiogenesis-related pathways, while GNA14 c.614A>T (p.Gln205Leu) mutation led to MAPK and angiogenesis-related pathway upregulation. Using a mouse xenograft model, we observed enlarged vessels from these mutations. Additionally, we initiated rapamycin treatment in a 14-year-old individual harboring the GNAQ c.626A>G (p.Gln209Arg) variant, resulting in gradual regression of cutaneous cavernous hemangiomas and improved motor strength, with minimal side effects. Understanding these mutations and their pathways provides a foundation for developing therapies for ECHs resistant to current therapies. Indeed, the administration of rapamycin in an individual within this study highlights the promise of targeted treatments in treating these complex lesions.
Collapse
Affiliation(s)
- Jian Ren
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Ziwei Cui
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Chendan Jiang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Leiming Wang
- Department of Pathology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yunqian Guan
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Yeqing Ren
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Shikun Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Tianqi Tu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Jiaxing Yu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Ye Li
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Wanru Duan
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Jian Guan
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Kai Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China
| | - Hongdian Zhang
- Department of Neurosurgery, The Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Dong Xing
- Biomedical Pioneering Innovation Center (BIOPIC), School of Life Sciences, Peking University, Beijing, China; Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China
| | - Mark L Kahn
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Hongqi Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China.
| | - Tao Hong
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, China International Neuroscience Institute, National Center for Neurological Disorders, Beijing, China; Department of Neurosurgery, Xiongan Xuanwu Hospital, Xiong'an New Area, China.
| |
Collapse
|
5
|
Saito A, Kise R, Inoue A. Generation of Comprehensive GPCR-Transducer-Deficient Cell Lines to Dissect the Complexity of GPCR Signaling. Pharmacol Rev 2024; 76:599-619. [PMID: 38719480 DOI: 10.1124/pharmrev.124.001186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/02/2024] [Accepted: 05/06/2024] [Indexed: 06/16/2024] Open
Abstract
G-protein-coupled receptors (GPCRs) compose the largest family of transmembrane receptors and are targets of approximately one-third of Food and Drug Administration-approved drugs owing to their involvement in almost all physiologic processes. GPCR signaling occurs through the activation of heterotrimeric G-protein complexes and β-arrestins, both of which serve as transducers, resulting in distinct cellular responses. Despite seeming simple at first glance, accumulating evidence indicates that activation of either transducer is not a straightforward process as a stimulation of a single molecule has the potential to activate multiple signaling branches. The complexity of GPCR signaling arises from the aspects of G-protein-coupling selectivity, biased signaling, interpathway crosstalk, and variable molecular modifications generating these diverse signaling patterns. Numerous questions relative to these aspects of signaling remained unanswered until the recent development of CRISPR genome-editing technology. Such genome editing technology presents opportunities to chronically eliminate the expression of G-protein subunits, β-arrestins, G-protein-coupled receptor kinases (GRKs), and many other signaling nodes in the GPCR pathways at one's convenience. Here, we review the practicality of using CRISPR-derived knockout (KO) cells in the experimental contexts of unraveling the molecular details of GPCR signaling mechanisms. To mention a few, KO cells have revealed the contribution of β-arrestins in ERK activation, Gα protein selectivity, GRK-based regulation of GPCRs, and many more, hence validating its broad applicability in GPCR studies. SIGNIFICANCE STATEMENT: This review emphasizes the practical application of G-protein-coupled receptor (GPCR) transducer knockout (KO) cells in dissecting the intricate regulatory mechanisms of the GPCR signaling network. Currently available cell lines, along with accumulating KO cell lines in diverse cell types, offer valuable resources for systematically elucidating GPCR signaling regulation. Given the association of GPCR signaling with numerous diseases, uncovering the system-based signaling map is crucial for advancing the development of novel drugs targeting specific diseases.
Collapse
Affiliation(s)
- Ayaki Saito
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ryoji Kise
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
6
|
Han X, Pinto LG, Vilar B, McNaughton PA. Opioid-Induced Hyperalgesia and Tolerance Are Driven by HCN Ion Channels. J Neurosci 2024; 44:e1368232023. [PMID: 38124021 PMCID: PMC11059424 DOI: 10.1523/jneurosci.1368-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/24/2023] [Accepted: 11/19/2023] [Indexed: 12/23/2023] Open
Abstract
Prolonged exposure to opioids causes an enhanced sensitivity to painful stimuli (opioid-induced hyperalgesia, OIH) and a need for increased opioid doses to maintain analgesia (opioid-induced tolerance, OIT), but the mechanisms underlying both processes remain obscure. We found that pharmacological block or genetic deletion of HCN2 ion channels in primary nociceptive neurons of male mice completely abolished OIH but had no effect on OIT. Conversely, pharmacological inhibition of central HCN channels alleviated OIT but had no effect on OIH. Expression of C-FOS, a marker of neuronal activity, was increased in second-order neurons of the dorsal spinal cord by induction of OIH, and the increase was prevented by peripheral block or genetic deletion of HCN2, but block of OIT by spinal block of HCN channels had no impact on C-FOS expression in dorsal horn neurons. Collectively, these observations show that OIH is driven by HCN2 ion channels in peripheral nociceptors, while OIT is driven by a member of the HCN family located in the CNS. Induction of OIH increased cAMP in nociceptive neurons, and a consequent shift in the activation curve of HCN2 caused an increase in nociceptor firing. The shift in HCN2 was caused by expression of a constitutively active μ-opioid receptor (MOR) and was reversed by MOR antagonists. We identified the opioid-induced MOR as a six-transmembrane splice variant, and we show that it increases cAMP by coupling constitutively to Gs HCN2 ion channels therefore drive OIH, and likely OIT, and may be a novel therapeutic target for the treatment of addiction.
Collapse
Affiliation(s)
- Xue Han
- Wolfson Sensory, Pain and Regeneration Centre, King's College London, London SE1 1UL, United Kingdom
| | - Larissa Garcia Pinto
- Wolfson Sensory, Pain and Regeneration Centre, King's College London, London SE1 1UL, United Kingdom
| | - Bruno Vilar
- Wolfson Sensory, Pain and Regeneration Centre, King's College London, London SE1 1UL, United Kingdom
| | - Peter A McNaughton
- Wolfson Sensory, Pain and Regeneration Centre, King's College London, London SE1 1UL, United Kingdom
| |
Collapse
|
7
|
Voss JH. Recommended Tool Compounds: Application of YM-254890 and FR900359 to Interrogate Gα q/11-Mediated Signaling Pathways. ACS Pharmacol Transl Sci 2023; 6:1790-1800. [PMID: 38093837 PMCID: PMC10714435 DOI: 10.1021/acsptsci.3c00214] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/16/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2024]
Abstract
The macrocyclic depsipeptides YM-254890 (YM) and FR900359 (FR) are natural products, which inhibit heterotrimeric Gαq/11 proteins with high potency and outstanding selectivity. Historically, pharmacological modulation of Gα proteins was only achieved by treatment with pertussis toxin and cholera toxin, whose application can be tedious and is restricted to the inhibition of Gαi/o proteins and activation of Gαs proteins, respectively. The breakthrough discovery and characterization of YM and FR rendered the closely related Gαq, Gα11, and Gα14 proteins amenable to pharmacological inhibition, and since then, both compounds have become widely used in molecular pharmacology and were also proven to be efficacious in animal models of disease. In the past years, both YM and FR were thoroughly characterized and have substantially contributed to an improved understanding of Gαq/11 signaling on a molecular and cellular level. Yet, the possibilities to interrogate Gαq/11 signaling in complex systems have only been exploited in a very limited number of studies, whose promising initial results warrant further application of YM and FR in basic and translational research. As both compounds have become commercially available as of late, this review focuses on their application in cell-based assays and in vivo systems, highlighting their qualities as tool compounds and providing instructions for their use.
Collapse
Affiliation(s)
- Jan Hendrik Voss
- Department of Physiology and Pharmacology,
Section of Receptor Biology and Signaling, Karolinska Institutet, S-171 65 Stockholm, Sweden
| |
Collapse
|
8
|
Kamato D, Gabr M, Kumarapperuma H, Chia ZJ, Zheng W, Xu S, Osman N, Little PJ. Gαq Is the Specific Mediator of PAR-1 Transactivation of Kinase Receptors in Vascular Smooth Muscle Cells. Int J Mol Sci 2022; 23:ijms232214425. [PMID: 36430902 PMCID: PMC9692893 DOI: 10.3390/ijms232214425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022] Open
Abstract
AIMS G protein-coupled receptor (GPCR) transactivation of kinase receptors greatly expands the actions attributable to GPCRs. Thrombin, via its cognate GPCR, protease-activated receptor (PAR)-1, transactivates tyrosine and serine/threonine kinase receptors, specifically the epidermal growth factor receptor and transforming growth factor-β receptor, respectively. PAR-1 transactivation-dependent signalling leads to the modification of lipid-binding proteoglycans involved in the retention of lipids and the development of atherosclerosis. The mechanisms of GPCR transactivation of kinase receptors are distinct. We aimed to investigate the role of proximal G proteins in transactivation-dependent signalling. MAIN METHODS Using pharmacological and molecular approaches, we studied the role of the G⍺ subunits, G⍺q and G⍺11, in the context of PAR-1 transactivation-dependent signalling leading to proteoglycan modifications. KEY FINDINGS Pan G⍺q subunit inhibitor UBO-QIC/FR900359 inhibited PAR-1 transactivation of kinase receptors and proteoglycans modification. The G⍺q/11 inhibitor YM254890 did not affect PAR-1 transactivation pathways. Molecular approaches revealed that of the two highly homogenous G⍺q members, G⍺q and G⍺11, only the G⍺q was involved in regulating PAR-1 mediated proteoglycan modification. Although G⍺q and G⍺11 share approximately 90% homology at the protein level, we show that the two isoforms exhibit different functional roles. SIGNIFICANCE Our findings may be extrapolated to other GPCRs involved in vascular pathology and highlight the need for novel pharmacological tools to assess the role of G proteins in GPCR signalling to expand the preeminent position of GPCRs in human therapeutics.
Collapse
Affiliation(s)
- Danielle Kamato
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
- School of Environment and Science, Griffith University, Nathan, QLD 4111, Australia
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia
- Correspondence:
| | - Mai Gabr
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Hirushi Kumarapperuma
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Zheng J. Chia
- Discovery Biology, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Wenhua Zheng
- Centre of Reproduction, Development & Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau 999078, China
| | - Suowen Xu
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of University of Science and Technology of China, Hefei 230052, China
| | - Narin Osman
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083, Australia
| | - Peter J. Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou 510520, China
- Sunshine Coast Health Institute, University of the Sunshine Coast, Birtinya, QLD 4575, Australia
| |
Collapse
|
9
|
Voss JH, Nagel J, Rafehi M, Guixà-González R, Malfacini D, Patt J, Kehraus S, Inoue A, König GM, Kostenis E, Deupi X, Namasivayam V, Müller CE. Unraveling binding mechanism and kinetics of macrocyclic Gα q protein inhibitors. Pharmacol Res 2021; 173:105880. [PMID: 34506902 DOI: 10.1016/j.phrs.2021.105880] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/23/2021] [Accepted: 09/05/2021] [Indexed: 10/20/2022]
Abstract
G proteins represent intracellular switches that transduce signals relayed from G protein-coupled receptors. The structurally related macrocyclic depsipeptides FR900359 (FR) and YM-254890 (YM) are potent, selective inhibitors of the Gαq protein family. We recently discovered that radiolabeled FR and YM display strongly divergent residence times, which translates into significantly longer antiasthmatic effects of FR. The present study is aimed at investigating the molecular basis for this observed disparity. Based on docking studies, we mutated amino acid residues of the Gαq protein predicted to interact with FR or YM, and recombinantly expressed the mutated Gαq proteins in cells in which the native Gαq proteins had been knocked out by CRISPR-Cas9. Both radioligands showed similar association kinetics, and their binding followed a conformational selection mechanism, which was rationalized by molecular dynamics simulation studies. Several mutations of amino acid residues near the putative binding site of the "lipophilic anchors" of FR, especially those predicted to interact with the isopropyl group present in FR but not in YM, led to dramatically accelerated dissociation kinetics. Our data indicate that the long residence time of FR depends on lipophilic interactions within its binding site. The observed structure-kinetic relationships point to a complex binding mechanism of FR, which likely involves snap-lock- or dowel-like conformational changes of either ligand or protein, or both. These experimental data will be useful for the design of compounds with a desired residence time, a parameter that has now been recognized to be of utmost importance in drug development.
Collapse
Affiliation(s)
- Jan H Voss
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Jessica Nagel
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Muhammad Rafehi
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Ramon Guixà-González
- Condensed Matter Theory Group, Paul Scherrer Institute (PSI), Forschungsstrasse 111, Villigen 5232, Switzerland
| | - Davide Malfacini
- Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53113 Bonn, Germany
| | - Julian Patt
- Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53113 Bonn, Germany
| | - Stefan Kehraus
- Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53113 Bonn, Germany
| | - Asuka Inoue
- Tohoku University, Graduate School of Pharmaceutical Sciences, Sendai, Miyagi 980-8578 Japan
| | - Gabriele M König
- Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53113 Bonn, Germany
| | - Evi Kostenis
- Institute of Pharmaceutical Biology, University of Bonn, Nussallee 6, 53113 Bonn, Germany
| | - Xavier Deupi
- Condensed Matter Theory Group, Paul Scherrer Institute (PSI), Forschungsstrasse 111, Villigen 5232, Switzerland; Laboratory of Biomolecular Research, Paul Scherrer Institute (PSI), Forschungsstrasse 111, Villigen 5232, Switzerland
| | - Vigneshwaran Namasivayam
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, D-53121 Bonn, Germany.
| |
Collapse
|
10
|
Hermes C, König GM, Crüsemann M. The chromodepsins - chemistry, biology and biosynthesis of a selective Gq inhibitor natural product family. Nat Prod Rep 2021; 38:2276-2292. [PMID: 33998635 DOI: 10.1039/d1np00005e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Covering: up to April 2021The bacterial cyclic depsipeptides FR900359 (FR) and YM-254890 (YM) were shown to selectively inhibit Gαq proteins with high potency and selectivity and have recently emerged as valuable pharmacological tools due to their effective mechanism of action. Here, we summarize important aspects of this small and specialized natural product family, for which we propose the name chromodepsins, starting from their discovery, producing organisms and structural variety. We then review biosynthesis, structure-activity relationships and ecological and evolutionary aspects of the chromodepsins. Lastly, we discuss their mechanism of action, potential medicinal applications and future opportunities and challenges for further use and development of these complex inhibitor molecules from nature.
Collapse
Affiliation(s)
- Cornelia Hermes
- Institute of Pharmaceutical Biology, Rheinische Friedrich-Wilhelms-University of Bonn, 53115 Bonn, Germany.
| | - Gabriele M König
- Institute of Pharmaceutical Biology, Rheinische Friedrich-Wilhelms-University of Bonn, 53115 Bonn, Germany.
| | - Max Crüsemann
- Institute of Pharmaceutical Biology, Rheinische Friedrich-Wilhelms-University of Bonn, 53115 Bonn, Germany.
| |
Collapse
|
11
|
Patt J, Alenfelder J, Pfeil EM, Voss JH, Merten N, Eryilmaz F, Heycke N, Rick U, Inoue A, Kehraus S, Deupi X, Müller CE, König GM, Crüsemann M, Kostenis E. An experimental strategy to probe Gq contribution to signal transduction in living cells. J Biol Chem 2021; 296:100472. [PMID: 33639168 PMCID: PMC8024710 DOI: 10.1016/j.jbc.2021.100472] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022] Open
Abstract
Heterotrimeric G protein subunits Gαq and Gα11 are inhibited by two cyclic depsipeptides, FR900359 (FR) and YM-254890 (YM), both of which are being used widely to implicate Gq/11 proteins in the regulation of diverse biological processes. An emerging major research question therefore is whether the cellular effects of both inhibitors are on-target, that is, mediated via specific inhibition of Gq/11 proteins, or off-target, that is, the result of nonspecific interactions with other proteins. Here we introduce a versatile experimental strategy to discriminate between these possibilities. We developed a Gαq variant with preserved catalytic activity, but refractory to FR/YM inhibition. A minimum of two amino acid changes were required and sufficient to achieve complete inhibitor resistance. We characterized the novel mutant in HEK293 cells depleted by CRISPR–Cas9 of endogenous Gαq and Gα11 to ensure precise control over the Gα-dependent cellular signaling route. Using a battery of cellular outcomes with known and concealed Gq contribution, we found that FR/YM specifically inhibited cellular signals after Gαq introduction via transient transfection. Conversely, both inhibitors were inert across all assays in cells expressing the drug-resistant variant. These findings eliminate the possibility that inhibition of non-Gq proteins contributes to the cellular effects of the two depsipeptides. We conclude that combined application of FR or YM along with the drug-resistant Gαq variant is a powerful in vitro strategy to discern on-target Gq against off-target non-Gq action. Consequently, it should be of high value for uncovering Gq input to complex biological processes with high accuracy and the requisite specificity.
Collapse
Affiliation(s)
- Julian Patt
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Judith Alenfelder
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Eva Marie Pfeil
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Jan Hendrik Voss
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Nicole Merten
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Funda Eryilmaz
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Nina Heycke
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Uli Rick
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Stefan Kehraus
- Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Xavier Deupi
- Laboratory of Biomolecular Research and Condensed Matter Theory Group, Paul Scherrer Institute, Villigen, Switzerland
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Gabriele M König
- Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Max Crüsemann
- Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany.
| |
Collapse
|
12
|
Schlegel JG, Tahoun M, Seidinger A, Voss JH, Kuschak M, Kehraus S, Schneider M, Matthey M, Fleischmann BK, König GM, Wenzel D, Müller CE. Macrocyclic Gq Protein Inhibitors FR900359 and/or YM-254890-Fit for Translation? ACS Pharmacol Transl Sci 2021; 4:888-897. [PMID: 33860209 DOI: 10.1021/acsptsci.1c00021] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Indexed: 12/11/2022]
Abstract
Guanine nucleotide-binding proteins (G proteins) transduce extracellular signals received by G protein-coupled receptors (GPCRs) to intracellular signaling cascades. While GPCRs represent the largest class of drug targets, G protein inhibition has only recently been recognized as a novel strategy for treating complex diseases such as asthma, inflammation, and cancer. The structurally similar macrocyclic depsipeptides FR900359 (FR) and YM-254890 (YM) are potent selective inhibitors of the Gq subfamily of G proteins. FR and YM differ in two positions, FR being more lipophilic than YM. Both compounds are utilized as pharmacological tools to block Gq proteins in vitro and in vivo. However, no detailed characterization of FR and YM has been performed, which is a prerequisite for the compounds' translation into clinical application. Here, we performed a thorough study of both compounds' physicochemical, pharmacokinetic, and pharmacological properties. Chemical stability was high across a large range of pH values, with FR being somewhat more stable than YM. Oral bioavailability and brain penetration of both depsipeptides were low. FR showed lower plasma protein binding and was metabolized significantly faster than YM by human and mouse liver microsomes. FR accumulated in lung after chronic intratracheal or intraperitoneal application, while YM was more distributed to other organs. Most strikingly, the previously observed longer residence time of FR resulted in a significantly prolonged pharmacologic effect as compared to YM in a methacholine-induced bronchoconstriction mouse model. These results prove that changes within a molecule which seem marginal compared to its structural complexity can lead to crucial pharmacological differences.
Collapse
Affiliation(s)
- Jonathan G Schlegel
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Mariam Tahoun
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Alexander Seidinger
- Department of Systems Physiology, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany
| | - Jan H Voss
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Markus Kuschak
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Stefan Kehraus
- Institute for Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany
| | - Marion Schneider
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Michaela Matthey
- Department of Systems Physiology, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany
| | - Bernd K Fleischmann
- Institute of Physiology I, Life & Brain Center, Medical Faculty, University of Bonn, 53105 Bonn, Germany
| | - Gabriele M König
- Institute for Pharmaceutical Biology, University of Bonn, 53115 Bonn, Germany
| | - Daniela Wenzel
- Department of Systems Physiology, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany.,Institute of Physiology I, Life & Brain Center, Medical Faculty, University of Bonn, 53105 Bonn, Germany
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| |
Collapse
|
13
|
Song G, Zhu X, Xuan Z, Zhao L, Dong H, Chen J, Li Z, Song W, Jin C, Zhou M, Xie H, Zheng S, Song P. Hypermethylation of GNA14 and its tumor-suppressive role in hepatitis B virus-related hepatocellular carcinoma. Am J Cancer Res 2021; 11:2318-2333. [PMID: 33500727 PMCID: PMC7797690 DOI: 10.7150/thno.48739] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 11/14/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal cancers worldwide, and its specific mechanism has not been fully elucidated. Inactivation of tumor suppressors may contribute to the occurrence, progression, and recurrence of HCC. DNA methylation is a crucial mechanism involved in regulating the occurrence of HCC. Herein, we aimed to identify the key methylation-related tumor suppressors as well as potential biomarkers and therapeutic targets in HCC. Methods: Combined analysis of TCGA and GEO databases was performed to obtain potential methylation-related tumor suppressors in HCC. Methyl-target sequencing was performed to analyze the methylation level of the GNA14 promoter. The diagnostic value of GNA14 as a predictor of HCC was evaluated in HCC tumor samples and compared with normal tissues. The functional role of GNA14 and its upstream and downstream regulatory factors were investigated by gain-of-function and loss-of-function assays in vitro. Subcutaneous tumorigenesis, lung colonization, and orthotopic liver tumor model were performed to analyze the role of GNA14 in vivo. Results: The expression of GNA14 was found to be downregulated in HCC and it was negatively correlated with hepatitis B virus (HBV) infection, vascular invasion, and prognosis of HCC. DNA methylation was demonstrated to be responsible for the altered expression of GNA14 and was regulated by HBV-encoded X protein (HBx). GNA14 regulated the RB pathway by promoting Notch1 cleavage to inhibit tumor proliferation, and might inhibit tumor metastasis by inhibiting the expression of JMJD6. Conclusion: GNA14 could be regulated by HBx by modulating the methylation status of its promoter. We identified GNA14 as a potential biomarker and therapeutic target for HCC.
Collapse
|