1
|
Löbbert A, Lorz N, Matthees ESF, Rößler P, Hoffmann C, Gossert AD. GPCR kinases phosphorylate GPCR C-terminal peptides in a hierarchical manner. Commun Biol 2025; 8:899. [PMID: 40490497 DOI: 10.1038/s42003-025-08301-7] [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: 04/26/2025] [Accepted: 05/27/2025] [Indexed: 06/11/2025] Open
Abstract
Responses from G protein-coupled receptors (GPCRs) are downregulated in a precisely orchestrated process called desensitization. This process consists of two major steps: phosphorylation of the receptor by GPCR kinases (GRKs), predominantly on its C-terminus, and recruitment of arrestin, resulting in different signaling outcomes. Yet, it remains unclear how the phosphorylation pattern on the receptor is determined. We carried out an NMR-based study of the phosphorylation patterns generated by GRK1 and GRK2 on C-terminal peptides of selected receptors (rhodopsin for GRK1, and β1- and β2-adrenergic receptors (ARs) for GRK2). Our data reveal that the kinases are promiscuous with respect to the substrate peptide, but produce clearly defined phosphorylation patterns on each substrate. We found pronounced differences in the rates at which certain residues are phosphorylated, in particular in the PXPP motifs in rhodopsin and β1AR. These results show that GRKs produce well-defined phosphorylation patterns in absence of further modulators like the full receptor or Gβγ, and that the time profile of the phosphorylation barcode seems to be largely encoded in the minimal pair of C-terminal peptide and GRK. The data further suggest that arrestin might encounter different phosphorylation barcodes over time, hinting at the possibility of time-dependent arrestin responses.
Collapse
Affiliation(s)
- Arnelle Löbbert
- Institute of Biochemistry, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Nils Lorz
- Institute of Biochemistry, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Edda S F Matthees
- Institute of Molecular Cell Biology, University Hospital Jena, Friedrich Schiller University, Jena, Germany
| | - Philip Rößler
- Institute of Biochemistry, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Carsten Hoffmann
- Institute of Molecular Cell Biology, University Hospital Jena, Friedrich Schiller University, Jena, Germany.
| | - Alvar D Gossert
- Institute of Biochemistry, Department of Biology, ETH Zürich, Zürich, Switzerland.
| |
Collapse
|
2
|
Janetzko J, Deutsch J, Shi Y, Siepe DH, Masureel M, Liu W, Viner R, Inoue A, Kobilka BK, Shivnaraine RV. Membrane phosphoinositides allosterically tune β-arrestin dynamics to facilitate GPCR core engagement. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.06.06.658200. [PMID: 40501946 PMCID: PMC12157425 DOI: 10.1101/2025.06.06.658200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/29/2025]
Abstract
Arrestin proteins bind active G protein-coupled receptors (GPCRs) through coordinated protein-protein, protein-phosphate, and protein-lipid interactions to attenuate G protein signaling and promote GPCR internalization and trafficking. While there are hundreds of diverse GPCRs, only two β-arrestin isoforms (βarrs) must recognize and engage this wide range of receptors with varied phosphorylation patterns. Traditional models suggest that βarr activation requires displacement of its autoinhibitory C-tail by a phosphorylated GPCR C-terminus; however, this paradigm fails to explain how minimally phosphorylated GPCRs still complex with βarrs. Using single-molecule Förster resonance energy transfer imaging and hydrogen-deuterium exchange mass spectrometry, we observe basal dynamics in which the βarr1 C-tail spontaneously releases from the N-domain, transiently adopting an active conformation that can facilitate binding of the phosphorylated GPCR C-terminus. We further demonstrate the importance of an intermediate state of βarr1 arising from spontaneous C-tail release stabilized by the membrane phosphoinositide PI(4,5)P2. Both PI(4,5)P2 and mutations in the proximal or middle regions of the C-tail shift βarr1 towards a partially released state, revealing an allosteric connection that informs a refined model for βarr activation. In this model, membrane engagement conformationally primes βarrs prior to receptor binding, thereby explaining how βarrs are recruited by diverse GPCRs, even those with limited C-terminal phosphorylation.
Collapse
Affiliation(s)
- John Janetzko
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
- Present address: Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Present address: Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Present address: Department of Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Present address: BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Jonathan Deutsch
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - Yuqi Shi
- ThermoFisher Scientific, San Jose, CA, USA
| | | | - Matthieu Masureel
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
- Present address: Department of Structural Biology, Genentech Inc., San Francisco, CA, USA
| | | | - Rosa Viner
- ThermoFisher Scientific, San Jose, CA, USA
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
- Department of Molecular Pharmacology, Kyoto University, Japan
| | - Brian K. Kobilka
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
| | - Rabindra V. Shivnaraine
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA
- Present address: Greenstone Biosciences, Palo Alto, CA, USA
| |
Collapse
|
3
|
Wang S, Kundu D, Zhang X, Tian X, Peng L, Kim KM. Mdm2-Mediated Ubiquitination Plays a Pivotal Role in Differentiating the Endocytic Roles of GRK2 and Arrestin3. Int J Mol Sci 2025; 26:3238. [PMID: 40244017 PMCID: PMC11989279 DOI: 10.3390/ijms26073238] [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: 02/19/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
Upon activation of certain G protein-coupled receptors, Mdm2 promotes the ubiquitination of both GRK2 and arrestin3. Similar to arrestin3, GRK2 ubiquitination was associated with its endocytic activity and proteasomal degradation. Ubiquitination of GRK2 was essential for arrestin3 ubiquitination, and vice versa. Cellular components involved in arrestin3 ubiquitination, including Gβγ, clathrin, and 14-3-3η, were also necessary for GRK2 ubiquitination. Additionally, the arrestin-biased signaling pathway contributed to the ubiquitination of both GRK2 and arrestin3. By employing Mdm2-knockdown cells alongside GRK2 and arrestin3 mutants deficient in ubiquitination sites, as well as receptors lacking phosphorylation sites, we established that the ubiquitinated forms of GRK2 and arrestin3 facilitate clathrin-dependent endocytosis, whereas non-ubiquitinated GRK2 and arrestin3 are responsible for caveolar and a distinct third endocytic pathway, respectively. In the context of clathrin-mediated endocytosis, arrestin3's interaction with clathrin and GRK2's interaction with the β2-adaptin subunit of adaptor protein complex 2 were critical. These findings suggest that GRK2 and arrestin3 ubiquitination are mutually dependent, with their ubiquitination states determining their roles in distinct endocytic pathways.
Collapse
Affiliation(s)
- Shujie Wang
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 61186, Republic of Korea; (S.W.); (D.K.); (X.T.); (L.P.)
| | - Dooti Kundu
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 61186, Republic of Korea; (S.W.); (D.K.); (X.T.); (L.P.)
| | - Xiaohan Zhang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, China;
| | - Xinru Tian
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 61186, Republic of Korea; (S.W.); (D.K.); (X.T.); (L.P.)
| | - Lulu Peng
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 61186, Republic of Korea; (S.W.); (D.K.); (X.T.); (L.P.)
| | - Kyeong-Man Kim
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 61186, Republic of Korea; (S.W.); (D.K.); (X.T.); (L.P.)
| |
Collapse
|
4
|
Millette MA, Coutinho A, Prieto M, Salesse C. Role of the Palmitoyl Group and of the Amphipathic α Helix in the Membrane Binding of the C-Terminus of G-Protein Receptor Kinase 4α/β. Biochemistry 2025; 64:987-1005. [PMID: 39977231 DOI: 10.1021/acs.biochem.4c00492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Membrane binding of monotopic proteins can involve various post-translational modifications or a combination of some membrane-binding elements. For example, amphipathic α helices and palmitoylation could drive the membrane attachment of proteins. G-protein-coupled receptor kinases (GRKs) regulate the activity of G-protein-coupled receptors. Several members of the family of GRKs are acylated. Moreover, the C-terminus of GRK6 contains an amphipathic α helix and a palmitoyl group, which could also be the case for GRK4 isoforms. In our experiments, GRK4α/β-derived peptides of differing C-terminal lengths (Cter-GRK4α/β variants) were thus studied to discriminate the individual role of the palmitoyl group and amphipathic α helix of Cter-GRK4α/β in its membrane binding. The membrane binding of the Cter-GRK4α/β variants was studied by comparing their maximum insertion pressure (MIP) to lipid monolayers as well as their intrinsic fluorescence properties using large unilamellar vesicles. The MIP data show a higher level of binding of the palmitoylated longest GRK4α/β variant. Moreover, MIP measurements in the absence and presence of 15 mol % of the negatively charged phosphoserine demonstrated that the amphipathic α helix of Cter-GRK4α/β plays a major role in its membrane binding. Accordingly, partition studies of the Cter-GRK4α/β variants to membranes by fluorescence spectroscopy demonstrate the involvement of the palmitoyl group and the amphipathic α helix of the C-terminus of GRK4α/β in its membrane binding. Altogether, the data show that both the palmitoyl group and the amphipathic helix highly favor membrane binding of the C-terminus of GRK4α/β, which should facilitate the proper anchoring of GRK4α/β and phosphorylation of GPCRs.
Collapse
Affiliation(s)
- Marc-Antoine Millette
- CUO-Recherche, Centre de recherche du CHU de Québec and Département d'ophtalmologie, Faculté de médecine, and Regroupement stratégique PROTEO, Université Laval, Québec, Québec G1V 0A6, Canada
| | - Ana Coutinho
- Centro de Química Estrutural, Institute of Molecular Sciences, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
- iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Manuel Prieto
- iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Christian Salesse
- CUO-Recherche, Centre de recherche du CHU de Québec and Département d'ophtalmologie, Faculté de médecine, and Regroupement stratégique PROTEO, Université Laval, Québec, Québec G1V 0A6, Canada
| |
Collapse
|
5
|
Kundu D, Min X, Zhang X, Tian X, Wang S, Kim KM. The Ubiquitination of Arrestin3 within the Nucleus Triggers the Nuclear Export of Mdm2, Which, in Turn, Mediates the Ubiquitination of GRK2 in the Cytosol. Int J Mol Sci 2024; 25:9644. [PMID: 39273591 PMCID: PMC11395016 DOI: 10.3390/ijms25179644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
GRK2 and arrestin3, key players in the functional regulation of G protein-coupled receptors (GPCRs), are ubiquitinated by Mdm2, a nuclear protein. The agonist-induced increase in arrestin3 ubiquitination occurs in the nucleus, underscoring the crucial role of its nuclear translocation in this process. The ubiquitination of arrestin3 occurs in the nucleus, highlighting the pivotal role of its nuclear translocation in this process. In contrast, GRK2 cannot translocate into the nucleus; thus, facilitation of the cytosolic translocation of nuclear Mdm2 is required to ubiquitinate GRK2 in the cytosol. Among the explored cellular components and processes, arrestin, Gβγ, clathrin, and receptor phosphorylation were found to be required for the nuclear import of arrestin3, the ubiquitination of arrestin3 in the nucleus, nuclear export of Mdm2, and the ubiquitination of GRK2 in the cytosol. In conclusion, our findings demonstrate that agonist-induced ubiquitination of arrestin3 in the nucleus is interconnected with cytosolic GRK2 ubiquitination.
Collapse
Affiliation(s)
| | | | | | | | | | - Kyeong-Man Kim
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 61186, Republic of Korea
| |
Collapse
|
6
|
Yaduvanshi S, Kumar V. Fungal alkaloid malbrancheamide reorients the lipid binding domain of GRK5. J Biomol Struct Dyn 2024:1-12. [PMID: 38661007 DOI: 10.1080/07391102.2024.2333987] [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: 11/10/2023] [Accepted: 03/16/2024] [Indexed: 04/26/2024]
Abstract
G protein-coupled receptors (GPCRs) are the largest group of receptors involved in various types of signaling. GPCR signaling is regulated via receptor phosphorylation by G protein-coupled receptor kinases 5 (GRK5). Calmodulin (CaM), a universal Ca2+ sensor, inhibits receptor phosphorylation by binding to GRK5. However, the inhibitor malbrancheamide (MBC), which binds at CaM C-lobe, allows for receptor phosphorylation. To understand the phosphorylation mechanism by GRK5, we carried out a MD simulation of the CaM/GRK5 complex in the presence and absence of the MBC inhibitor. The lipid binding domain (LBD) of GRK5 adopted different positions in the presence and absence of inhibitor. Furthermore, the inhibitor MBC restricted the movement of the N-lobe tether (NLT) loop, probably blocking the autophosphorylation of GRK5.
Collapse
Affiliation(s)
- Shivani Yaduvanshi
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| | - Veerendra Kumar
- Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, India
| |
Collapse
|
7
|
Schafer CT, Chen Q, Tesmer JJG, Handel TM. Atypical Chemokine Receptor 3 "Senses" CXC Chemokine Receptor 4 Activation Through GPCR Kinase Phosphorylation. Mol Pharmacol 2023; 104:174-186. [PMID: 37474305 PMCID: PMC11033958 DOI: 10.1124/molpharm.123.000710] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/22/2023] [Accepted: 06/30/2023] [Indexed: 07/22/2023] Open
Abstract
Atypical chemokine receptor 3 (ACKR3) is an arrestin-biased receptor that regulates extracellular chemokine levels through scavenging. The scavenging process restricts the availability of the chemokine agonist CXCL12 for the G protein-coupled receptor (GPCR) CXCR4 and requires phosphorylation of the ACKR3 C-terminus by GPCR kinases (GRKs). ACKR3 is phosphorylated by GRK2 and GRK5, but the mechanisms by which these kinases regulate the receptor are unresolved. Here we determined that GRK5 phosphorylation of ACKR3 results in more efficient chemokine scavenging and β-arrestin recruitment than phosphorylation by GRK2 in HEK293 cells. However, co-activation of CXCR4-enhanced ACKR3 phosphorylation by GRK2 through the liberation of Gβγ, an accessory protein required for efficient GRK2 activity. The results suggest that ACKR3 "senses" CXCR4 activation through a GRK2-dependent crosstalk mechanism, which enables CXCR4 to influence the efficiency of CXCL12 scavenging and β-arrestin recruitment to ACKR3. Surprisingly, we also found that despite the requirement for phosphorylation and the fact that most ligands promote β-arrestin recruitment, β-arrestins are dispensable for ACKR3 internalization and scavenging, suggesting a yet-to-be-determined function for these adapter proteins. Since ACKR3 is also a receptor for CXCL11 and opioid peptides, these data suggest that such crosstalk may also be operative in cells with CXCR3 and opioid receptor co-expression. Additionally, kinase-mediated receptor cross-regulation may be relevant to other atypical and G protein-coupled receptors that share common ligands. SIGNIFICANCE STATEMENT: The atypical receptor ACKR3 indirectly regulates CXCR4-mediated cell migration by scavenging their shared agonist CXCL12. Here, we show that scavenging and β-arrestin recruitment by ACKR3 are primarily dependent on phosphorylation by GRK5. However, we also show that CXCR4 co-activation enhances the contribution of GRK2 by liberating Gβγ. This phosphorylation crosstalk may represent a common feedback mechanism between atypical and G protein-coupled receptors with shared ligands for regulating the efficiency of scavenging or other atypical receptor functions.
Collapse
Affiliation(s)
- Christopher T Schafer
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (C.T.S., T.M.H.) and Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (Q.C., J.J.G.T.)
| | - Qiuyan Chen
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (C.T.S., T.M.H.) and Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (Q.C., J.J.G.T.)
| | - John J G Tesmer
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (C.T.S., T.M.H.) and Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (Q.C., J.J.G.T.)
| | - Tracy M Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (C.T.S., T.M.H.) and Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (Q.C., J.J.G.T.)
| |
Collapse
|
8
|
Schafer CT, Chen Q, Tesmer JJG, Handel TM. Atypical Chemokine Receptor 3 'Senses' CXC Chemokine Receptor 4 Activation Through GPCR Kinase Phosphorylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.25.530029. [PMID: 36865154 PMCID: PMC9980177 DOI: 10.1101/2023.02.25.530029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2023]
Abstract
Atypical chemokine receptor 3 (ACKR3) is an arrestin-biased receptor that regulates extracellular chemokine levels through scavenging. The scavenging action mediates the availability of the chemokine CXCL12 for the G protein-coupled receptor (GPCR) CXCR4 and requires phosphorylation of the ACKR3 C-terminus by GPCR kinases (GRKs). ACKR3 is phosphorylated by GRK2 and GRK5, but the mechanisms by which these kinases regulate the receptor are unresolved. Here we mapped the phosphorylation patterns and determined that GRK5 phosphorylation of ACKR3 dominates β-arrestin recruitment and chemokine scavenging over GRK2. Co-activation of CXCR4 significantly enhanced phosphorylation by GRK2 through the liberation of Gβγ. These results suggest that ACKR3 'senses' CXCR4 activation through a GRK2-dependent crosstalk mechanism. Surprisingly, we also found that despite the requirement for phosphorylation, and the fact that most ligands promote β-arrestin recruitment, β-arrestins are dispensable for ACKR3 internalization and scavenging, suggesting a yet to be determined function for these adapter proteins.
Collapse
Affiliation(s)
- Christopher T. Schafer
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, USA
| | - Qiuyan Chen
- Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
- Present address: Dept. of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - John J. G. Tesmer
- Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA
| | - Tracy M. Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, USA
| |
Collapse
|
9
|
Chen Q, Tesmer JJG. G protein-coupled receptor interactions with arrestins and GPCR kinases: The unresolved issue of signal bias. J Biol Chem 2022; 298:102279. [PMID: 35863432 PMCID: PMC9418498 DOI: 10.1016/j.jbc.2022.102279] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 07/05/2022] [Accepted: 07/08/2022] [Indexed: 12/25/2022] Open
Abstract
G protein-coupled receptor (GPCR) kinases (GRKs) and arrestins interact with agonist-bound GPCRs to promote receptor desensitization and downregulation. They also trigger signaling cascades distinct from those of heterotrimeric G proteins. Biased agonists for GPCRs that favor either heterotrimeric G protein or GRK/arrestin signaling are of profound pharmacological interest because they could usher in a new generation of drugs with greatly reduced side effects. One mechanism by which biased agonism might occur is by stabilizing receptor conformations that preferentially bind to GRKs and/or arrestins. In this review, we explore this idea by comparing structures of GPCRs bound to heterotrimeric G proteins with those of the same GPCRs in complex with arrestins and GRKs. The arrestin and GRK complexes all exhibit high conformational heterogeneity, which is likely a consequence of their unusual ability to adapt and bind to hundreds of different GPCRs. This dynamic behavior, along with the experimental tactics required to stabilize GPCR complexes for biophysical analysis, confounds these comparisons, but some possible molecular mechanisms of bias are beginning to emerge. We also examine if and how the recent structures advance our understanding of how arrestins parse the "phosphorylation barcodes" installed in the intracellular loops and tails of GPCRs by GRKs. In the future, structural analyses of arrestins in complex with intact receptors that have well-defined native phosphorylation barcodes, such as those installed by the two nonvisual subfamilies of GRKs, will be particularly illuminating.
Collapse
Affiliation(s)
- Qiuyan Chen
- Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, USA
| | - John J G Tesmer
- Departments of Biological Sciences and of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, USA.
| |
Collapse
|
10
|
Karmakar S, Klauda JB. Modeling the Membrane Binding Mechanism of a Lipid Transport Protein Osh4 to Single Membranes. Biophys J 2022; 121:1560-1575. [PMID: 35247338 PMCID: PMC9072576 DOI: 10.1016/j.bpj.2022.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/13/2021] [Accepted: 03/01/2022] [Indexed: 11/26/2022] Open
Abstract
All atom molecular dynamics simulations are utilized to unravel the binding mechanism of yeast oxysterol binding protein (Osh4) to model membranes with varying anionic lipid concentration using all-atom (AA) and the highly mobile membrane mimetic (HMMM) representations. For certain protein-lipid interactions, an improved force field description is used (CUFIX) to accurately describe lipid-protein electrostatic interactions. Our detailed computational studies have identified a single, β-crease orientated, membrane bound conformation of Osh4 for all anionic membranes. The penetration of the PHE-239 residue below the membrane phosphate plane is the characteristic signature of the membrane-bound state of Osh4. As the phenylalanine loop anchors itself deeply in the membrane; the other regions of the Osh4, namely, ALPS motif, ß6- ß7 loop, ß14- ß15 loop and ß16- ß17 loop, maximize their contact with the membrane. Furthermore, loose lipid packing and higher mobility of HMMM enables stronger association of ALPS motif with the membrane lipids through its hydrophobic surface and after the HMMM is converted to AA and equilibrated the binding is 2-3 times stronger compared to simulations started with the AA representation yielding the major importance of the ALPS motif to binding. Quantitative estimation of binding energy revealed that the phenylalanine loop plays a crucial role in stable membrane attachment of Osh4 and contributes significantly toward overall binding process. The CUFIX parameters provide a more balanced picture of hydrophobic and electrostatic interactions between the protein and the membrane which differs from our past work that showed salt bridges alone stabilized Osh4-membrane contact. Our study provides a comprehensive picture of the binding mechanism of Osh4 with model single membranes and thus, understanding of the initial interactions is important for elucidating the biological function of this protein to shuttle lipids between organelles.
Collapse
Affiliation(s)
| | - Jeffery B Klauda
- Department of Chemical and Biomolecular Engineering; Biophysics Graduate Program University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
11
|
Suo WZ. GRK5 Deficiency Causes Mild Cognitive Impairment due to Alzheimer's Disease. J Alzheimers Dis 2021; 85:1399-1410. [PMID: 34958040 DOI: 10.3233/jad-215379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Prevention of Alzheimer's disease (AD) is a high priority mission while searching for a disease modifying therapy for AD, a devastating major public health crisis. Clinical observations have identified a prodromal stage of AD for which the patients have mild cognitive impairment (MCI) though do not yet meet AD diagnostic criteria. As an identifiable transitional stage before the onset of AD, MCI should become the high priority target for AD prevention, assuming successful prevention of MCI and/or its conversion to AD also prevents the subsequent AD. By pulling this string, one demonstrated cause of amnestic MCI appears to be the deficiency of G protein-coupled receptor-5 (GRK5). The most compelling evidence is that GRK5 knockout (GRK5KO) mice naturally develop into aMCI during aging. Moreover, GRK5 deficiency was reported to occur during prodromal stage of AD in CRND8 transgenic mice. When a GRK5KO mouse was crossbred with Tg2576 Swedish amyloid precursor protein transgenic mouse, the resulted double transgenic GAP mice displayed exaggerated behavioral and pathological changes across the spectrum of AD pathogenesis. Therefore, the GRK5 deficiency possesses unique features and advantage to serve as a prophylactic therapeutic target for MCI due to AD.
Collapse
Affiliation(s)
- William Z Suo
- Laboratory for Alzheimer's Disease & Aging Research, VA Medical Center, Kansas City, MO, USA.,Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA.,The University of Kansas Alzheimer's Disease Center, Kansas City, KS, USA
| |
Collapse
|
12
|
Overduin M, Kervin TA. The phosphoinositide code is read by a plethora of protein domains. Expert Rev Proteomics 2021; 18:483-502. [PMID: 34351250 DOI: 10.1080/14789450.2021.1962302] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The proteins that decipher nucleic acid- and protein-based information are well known, however, those that read membrane-encoded information remain understudied. Here we report 70 different human, microbial and viral protein folds that recognize phosphoinositides (PIs), comprising the readers of a vast membrane code. AREAS COVERED Membrane recognition is best understood for FYVE, PH and PX domains, which exemplify hundreds of PI code readers. Comparable lipid interaction mechanisms may be mediated by kinases, adjacent C1 and C2 domains, trafficking arrestin, GAT and VHS modules, membrane-perturbing annexin, BAR, CHMP, ENTH, HEAT, syntaxin and Tubby helical bundles, multipurpose FERM, EH, MATH, PHD, PDZ, PROPPIN, PTB and SH2 domains, as well as systems that regulate receptors, GTPases and actin filaments, transfer lipids and assembled bacterial and viral particles. EXPERT OPINION The elucidation of how membranes are recognized has extended the genetic code to the PI code. Novel discoveries include PIP-stop and MET-stop residues to which phosphates and metabolites are attached to block phosphatidylinositol phosphate (PIP) recognition, memteins as functional membrane protein apparatuses, and lipidons as lipid "codons" recognized by membrane readers. At least 5% of the human proteome senses such membrane signals and allows eukaryotic organelles and pathogens to operate and replicate.
Collapse
Affiliation(s)
- Michael Overduin
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Troy A Kervin
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
13
|
Mechanistic diversity involved in the desensitization of G protein-coupled receptors. Arch Pharm Res 2021; 44:342-353. [PMID: 33761113 DOI: 10.1007/s12272-021-01320-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/14/2021] [Indexed: 01/14/2023]
Abstract
The desensitization of G protein-coupled receptors (GPCRs), which involves rapid loss of responsiveness due to repeated or chronic exposure to agonists, can occur through various mechanisms at different levels of signaling pathways. In this review, the mechanisms of GPCR desensitization are classified according to their occurrence at the receptor level and downstream to the receptor. The desensitization at the receptor level occurs in a phosphorylation-dependent manner, wherein the activated receptors are phosphorylated by GPCR kinases (GRKs), thereby increasing their affinities for arrestins. Arrestins bind to receptors through the cavity on the cytoplasmic region of heptahelical domains and interfere with the binding and activation of G-protein. Diverse mechanisms are involved in the desensitization that occurs downstream of the receptor. Some of these include the sequestration of G proteins, such as Gq and Gi/o by GRK2/3 and deubiquitinated arrestins, respectively. Mechanistically, GRK2/3 attenuates GPCR signaling by sequestering the Gα subunits of the Gq family and Gβγ via regulators of G protein signaling and pleckstrin homology domains, respectively. Moreover, studies on Gi/o-coupled D2-like receptors have reported that arrestins are deubiquitinated under desensitization condition and form a stable complex with Gβγ, thereby preventing them from coupling with Gα and the receptor, eventually leading to receptor signaling inhibition. Notably, the desensitization mechanism that involves arrestin deubiquitination is interesting; however, this is a new mechanism and needs to be explored further.
Collapse
|
14
|
The Open Question of How GPCRs Interact with GPCR Kinases (GRKs). Biomolecules 2021; 11:biom11030447. [PMID: 33802765 PMCID: PMC8002388 DOI: 10.3390/biom11030447] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/13/2021] [Accepted: 03/15/2021] [Indexed: 01/14/2023] Open
Abstract
G protein-coupled receptors (GPCRs), which regulate a vast number of eukaryotic processes, are desensitized by various mechanisms but, most importantly, by the GPCR kinases (GRKs). Ever since GRKs were first identified, investigators have sought to determine which structural features of GRKs are used to select for the agonist-bound states of GPCRs and how this binding event in turn enhances GRK catalytic activity. Despite a wealth of molecular information from high-resolution crystal structures of GRKs, the mechanisms driving activation have remained elusive, in part because the GRK N-terminus and active site tether region, previously proposed to serve as a receptor docking site and to be key to kinase domain closure, are often disordered or adopt inconsistent conformations. However, two recent studies have implicated other regions of GRKs as being involved in direct interactions with active GPCRs. Atomic resolution structures of GPCR–GRK complexes would help refine these models but are, so far, lacking. Here, we assess three distinct models for how GRKs recognize activated GPCRs, discuss limitations in the approaches used to generate them, and then experimentally test a hypothetical GPCR interaction site in GRK2 suggested by the two newest models.
Collapse
|
15
|
Lavington S, Watts A. Lipid nanoparticle technologies for the study of G protein-coupled receptors in lipid environments. Biophys Rev 2020; 12:10.1007/s12551-020-00775-5. [PMID: 33215301 PMCID: PMC7755959 DOI: 10.1007/s12551-020-00775-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are a large family of integral membrane proteins which conduct a wide range of biological roles and represent significant drug targets. Most biophysical and structural studies of GPCRs have been conducted on detergent-solubilised receptors, and it is clear that detergents can have detrimental effects on GPCR function. Simultaneously, there is increasing appreciation of roles for specific lipids in modulation of GPCR function. Lipid nanoparticles such as nanodiscs and styrene maleic acid lipid particles (SMALPs) offer opportunities to study integral membrane proteins in lipid environments, in a form that is soluble and amenable to structural and biophysical experiments. Here, we review the application of lipid nanoparticle technologies to the study of GPCRs, assessing the relative merits and limitations of each system. We highlight how these technologies can provide superior platforms to detergents for structural and biophysical studies of GPCRs and inform on roles for protein-lipid interactions in GPCR function.
Collapse
Affiliation(s)
- Steven Lavington
- Biochemistry Department, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK
| | - Anthony Watts
- Biochemistry Department, University of Oxford, South Parks Road, Oxford, OX1 3QU, UK.
| |
Collapse
|
16
|
Szlasa W, Zendran I, Zalesińska A, Tarek M, Kulbacka J. Lipid composition of the cancer cell membrane. J Bioenerg Biomembr 2020; 52:321-342. [PMID: 32715369 PMCID: PMC7520422 DOI: 10.1007/s10863-020-09846-4] [Citation(s) in RCA: 236] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/10/2020] [Indexed: 12/12/2022]
Abstract
Cancer cell possesses numerous adaptations to resist the immune system response and chemotherapy. One of the most significant properties of the neoplastic cells is the altered lipid metabolism, and consequently, the abnormal cell membrane composition. Like in the case of phosphatidylcholine, these changes result in the modulation of certain enzymes and accumulation of energetic material, which could be used for a higher proliferation rate. The changes are so prominent, that some lipids, such as phosphatidylserines, could even be considered as the cancer biomarkers. Additionally, some changes of biophysical properties of cell membranes lead to the higher resistance to chemotherapy, and finally to the disturbances in signalling pathways. Namely, the increased levels of certain lipids, like for instance phosphatidylserine, lead to the attenuation of the immune system response. Also, changes in lipid saturation prevent the cells from demanding conditions of the microenvironment. Particularly interesting is the significance of cell membrane cholesterol content in the modulation of metastasis. This review paper discusses the roles of each lipid type in cancer physiology. The review combined theoretical data with clinical studies to show novel therapeutic options concerning the modulation of cell membranes in oncology.
Collapse
Affiliation(s)
- Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, Wrocław, Poland
| | - Iga Zendran
- Faculty of Medicine, Wroclaw Medical University, Wrocław, Poland
| | | | - Mounir Tarek
- Université de Lorraine, CNRS, LPCT, F-54000, Nancy, France
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Wrocław, Poland.
| |
Collapse
|
17
|
Forster L, Grätz L, Mönnich D, Bernhardt G, Pockes S. A Split Luciferase Complementation Assay for the Quantification of β-Arrestin2 Recruitment to Dopamine D 2-Like Receptors. Int J Mol Sci 2020; 21:ijms21176103. [PMID: 32847148 PMCID: PMC7503597 DOI: 10.3390/ijms21176103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/12/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022] Open
Abstract
Investigations on functional selectivity of GPCR ligands have become increasingly important to identify compounds with a potentially more beneficial side effect profile. In order to discriminate between individual signaling pathways, the determination of β-arrestin2 recruitment, in addition to G-protein activation, is of great value. In this study, we established a sensitive split luciferase-based assay with the ability to quantify β-arrestin2 recruitment to D2long and D3 receptors and measure time-resolved β-arrestin2 recruitment to the D2long receptor after agonist stimulation. We were able to characterize several standard (inverse) agonists as well as antagonists at the D2longR and D3R subtypes, whereas for the D4.4R, no β-arrestin2 recruitment was detected, confirming previous reports. Extensive radioligand binding studies and comparisons with the respective wild-type receptors confirm that the attachment of the Emerald luciferase fragment to the receptors does not affect the integrity of the receptor proteins. Studies on the involvement of GRK2/3 and PKC on the β-arrestin recruitment to the D2longR and D3R, as well as at the D1R using different kinase inhibitors, showed that the assay could also contribute to the elucidation of signaling mechanisms. Its broad applicability, which provides concentration-dependent and kinetic information on receptor/β-arrestin2 interactions, renders this homogeneous assay a valuable method for the identification of biased agonists.
Collapse
Affiliation(s)
- Lisa Forster
- Correspondence: (L.F.); (S.P.); Tel.: +49-941-943-4796 (L.F.); +49-941-943-4825 (S.P.)
| | | | | | | | - Steffen Pockes
- Correspondence: (L.F.); (S.P.); Tel.: +49-941-943-4796 (L.F.); +49-941-943-4825 (S.P.)
| |
Collapse
|
18
|
Perturbation of the interactions of calmodulin with GRK5 using a natural product chemical probe. Proc Natl Acad Sci U S A 2019; 116:15895-15900. [PMID: 31337679 DOI: 10.1073/pnas.1818547116] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
G protein-coupled receptor (GPCR) kinases (GRKs) are responsible for initiating desensitization of activated GPCRs. GRK5 is potently inhibited by the calcium-sensing protein calmodulin (CaM), which leads to nuclear translocation of GRK5 and promotion of cardiac hypertrophy. Herein, we report the architecture of the Ca2+·CaM-GRK5 complex determined by small-angle X-ray scattering and negative-stain electron microscopy. Ca2+·CaM binds primarily to the small lobe of the kinase domain of GRK5 near elements critical for receptor interaction and membrane association, thereby inhibiting receptor phosphorylation while activating the kinase for phosphorylation of soluble substrates. To define the role of each lobe of Ca2+·CaM, we utilized the natural product malbrancheamide as a chemical probe to show that the C-terminal lobe of Ca2+·CaM regulates membrane binding while the N-terminal lobe regulates receptor phosphorylation and kinase domain activation. In cells, malbrancheamide attenuated GRK5 nuclear translocation and effectively blocked the hypertrophic response, demonstrating the utility of this natural product and its derivatives in probing Ca2+·CaM-dependent hypertrophy.
Collapse
|
19
|
Huang D, Cao L, Xiao L, Song JX, Zhang YJ, Zheng P, Zheng SG. Hypoxia induces actin cytoskeleton remodeling by regulating the binding of CAPZA1 to F-actin via PIP2 to drive EMT in hepatocellular carcinoma. Cancer Lett 2019; 448:117-127. [PMID: 30742939 DOI: 10.1016/j.canlet.2019.01.042] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/25/2019] [Accepted: 01/29/2019] [Indexed: 12/11/2022]
Abstract
Studies have shown that hypoxia can induce cytoskeletal injury and remodeling through the activation of the RhoA/ROCK signaling pathway by hypoxia-inducible factor-1α (HIF-1α). Our previous study confirmed that CAPZA1 can modulate EMT by regulating actin cytoskeleton remodeling. However, the relationship between HIF-1α and CAPZA1 has not been illustrated. Therefore, this study aimed to investigate the mechanism by which hypoxia induces the remodeling of the actin cytoskeleton by regulating CAPZA1 in hepatocellular carcinoma (HCC) cells. In the present study, we showed that the low expression of CAPZA1 promotes HCC cell invasion and migration in vitro and in vivo by regulating actin cytoskeleton remodeling to drive EMT. Furthermore, we found that the combination of PIP2 and CAPZA1 enables CAPZA1 to be released from the barbed end of F-actin, which in turn drives the remodeling of the actin cytoskeleton. Finally, we confirmed that hypoxia increases PIP2 levels and its binding to CAPZA1 in HCC cells via the HIF-1α/RhoA/ROCK1 pathway. Thus, CAPZA1 and PIP2 could be therapeutic targets to inhibit the invasion and migration promoted by hypoxia in HCC cells.
Collapse
Affiliation(s)
- Deng Huang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Li Cao
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Le Xiao
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; General Surgery Center, Chengdu Military General Hospital, Chengdu, Sichuan Province, 610083, China
| | - Ju-Xian Song
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Hepatobiliary Surgery, Libration Army No. 925 Hospital, Guiyang City, Guizhou Province, 550009, China
| | - Yu-Jun Zhang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ping Zheng
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Shu-Guo Zheng
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
20
|
Nogués L, Palacios-García J, Reglero C, Rivas V, Neves M, Ribas C, Penela P, Mayor F. G protein-coupled receptor kinases (GRKs) in tumorigenesis and cancer progression: GPCR regulators and signaling hubs. Semin Cancer Biol 2018; 48:78-90. [DOI: 10.1016/j.semcancer.2017.04.013] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/22/2017] [Accepted: 04/26/2017] [Indexed: 12/13/2022]
|
21
|
Structural and Functional Analysis of a β 2-Adrenergic Receptor Complex with GRK5. Cell 2017; 169:407-421.e16. [PMID: 28431242 DOI: 10.1016/j.cell.2017.03.047] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/10/2017] [Accepted: 03/28/2017] [Indexed: 12/31/2022]
Abstract
The phosphorylation of agonist-occupied G-protein-coupled receptors (GPCRs) by GPCR kinases (GRKs) functions to turn off G-protein signaling and turn on arrestin-mediated signaling. While a structural understanding of GPCR/G-protein and GPCR/arrestin complexes has emerged in recent years, the molecular architecture of a GPCR/GRK complex remains poorly defined. We used a comprehensive integrated approach of cross-linking, hydrogen-deuterium exchange mass spectrometry (MS), electron microscopy, mutagenesis, molecular dynamics simulations, and computational docking to analyze GRK5 interaction with the β2-adrenergic receptor (β2AR). These studies revealed a dynamic mechanism of complex formation that involves large conformational changes in the GRK5 RH/catalytic domain interface upon receptor binding. These changes facilitate contacts between intracellular loops 2 and 3 and the C terminus of the β2AR with the GRK5 RH bundle subdomain, membrane-binding surface, and kinase catalytic cleft, respectively. These studies significantly contribute to our understanding of the mechanism by which GRKs regulate the function of activated GPCRs. PAPERCLIP.
Collapse
|
22
|
Steury MD, McCabe LR, Parameswaran N. G Protein-Coupled Receptor Kinases in the Inflammatory Response and Signaling. Adv Immunol 2017; 136:227-277. [PMID: 28950947 DOI: 10.1016/bs.ai.2017.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
G protein-coupled receptor kinases (GRKs) are serine/threonine kinases that regulate a large and diverse class of G protein-coupled receptors (GPCRs). Through GRK phosphorylation and β-arrestin recruitment, GPCRs are desensitized and their signal terminated. Recent work on these kinases has expanded their role from canonical GPCR regulation to include noncanonical regulation of non-GPCR and nonreceptor substrates through phosphorylation as well as via scaffolding functions. Owing to these and other regulatory roles, GRKs have been shown to play a critical role in the outcome of a variety of physiological and pathophysiological processes including chemotaxis, signaling, migration, inflammatory gene expression, etc. This diverse set of functions for these proteins makes them popular targets for therapeutics. Role for these kinases in inflammation and inflammatory disease is an evolving area of research currently pursued in many laboratories. In this review, we describe the current state of knowledge on various GRKs pertaining to their role in inflammation and inflammatory diseases.
Collapse
Affiliation(s)
| | - Laura R McCabe
- Michigan State University, East Lansing, MI, United States
| | | |
Collapse
|
23
|
See Hoe LE, May LT, Headrick JP, Peart JN. Sarcolemmal dependence of cardiac protection and stress-resistance: roles in aged or diseased hearts. Br J Pharmacol 2016; 173:2966-91. [PMID: 27439627 DOI: 10.1111/bph.13552] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 06/27/2016] [Accepted: 06/28/2016] [Indexed: 12/25/2022] Open
Abstract
Disruption of the sarcolemmal membrane is a defining feature of oncotic death in cardiac ischaemia-reperfusion (I-R), and its molecular makeup not only fundamentally governs this process but also affects multiple determinants of both myocardial I-R injury and responsiveness to cardioprotective stimuli. Beyond the influences of membrane lipids on the cytoprotective (and death) receptors intimately embedded within this bilayer, myocardial ionic homeostasis, substrate metabolism, intercellular communication and electrical conduction are all sensitive to sarcolemmal makeup, and critical to outcomes from I-R. As will be outlined in this review, these crucial sarcolemmal dependencies may underlie not only the negative effects of age and common co-morbidities on myocardial ischaemic tolerance but also the on-going challenge of implementing efficacious cardioprotection in patients suffering accidental or surgically induced I-R. We review evidence for the involvement of sarcolemmal makeup changes in the impairment of stress-resistance and cardioprotection observed with ageing and highly prevalent co-morbid conditions including diabetes and hypercholesterolaemia. A greater understanding of membrane changes with age/disease, and the inter-dependences of ischaemic tolerance and cardioprotection on sarcolemmal makeup, can facilitate the development of strategies to preserve membrane integrity and cell viability, and advance the challenging goal of implementing efficacious 'cardioprotection' in clinically relevant patient cohorts. Linked Articles This article is part of a themed section on Molecular Pharmacology of G Protein-Coupled Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v173.20/issuetoc.
Collapse
Affiliation(s)
- Louise E See Hoe
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia.,Critical Care Research Group, The Prince Charles Hospital and The University of Queensland, Chermside, Queensland, Australia
| | - Lauren T May
- Monash Institute of Pharmaceutical Sciences, Monash University, Clayton, VIC, Australia
| | - John P Headrick
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia
| | - Jason N Peart
- Menzies Health Institute Queensland, Griffith University, Gold Coast, Australia.
| |
Collapse
|
24
|
G-protein-coupled receptor kinase 2 terminates G-protein-coupled receptor function in steroid hormone 20-hydroxyecdysone signaling. Sci Rep 2016; 6:29205. [PMID: 27412951 PMCID: PMC4944123 DOI: 10.1038/srep29205] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 06/16/2016] [Indexed: 12/26/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) transmit extracellular signals across the cell membrane. GPCR kinases (GRKs) desensitize GPCR signals in the cell membrane. However, the role and mechanism of GRKs in the desensitization of steroid hormone signaling are unclear. In this study, we propose that GRK2 is phosphorylated by protein kinase C (PKC) in response to induction by the steroid hormone 20-hydroxyecdysone (20E), which determines its translocation to the cell membrane of the lepidopteran Helicoverpa armigera. GRK2 protein expression is increased during the metamorphic stage because of induction by 20E. Knockdown of GRK2 in larvae causes accelerated pupation, an increase in 20E-response gene expression, and advanced apoptosis and metamorphosis. 20E induces translocation of GRK2 from the cytoplasm to the cell membrane via steroid hormone ecdysone-responsive GPCR (ErGPCR-2). GRK2 is phosphorylated by PKC on serine 680 after induction by 20E, which leads to the translocation of GRK2 to the cell membrane. GRK2 interacts with ErGPCR-2. These data indicate that GRK2 terminates the ErGPCR-2 function in 20E signaling in the cell membrane by a negative feedback mechanism.
Collapse
|
25
|
Hullmann J, Traynham CJ, Coleman RC, Koch WJ. The expanding GRK interactome: Implications in cardiovascular disease and potential for therapeutic development. Pharmacol Res 2016; 110:52-64. [PMID: 27180008 DOI: 10.1016/j.phrs.2016.05.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 05/05/2016] [Indexed: 12/11/2022]
Abstract
Heart failure (HF) is a global epidemic with the highest degree of mortality and morbidity of any disease presently studied. G protein-coupled receptors (GPCRs) are prominent regulators of cardiovascular function. Activated GPCRs are "turned off" by GPCR kinases (GRKs) in a process known as "desensitization". GRKs 2 and 5 are highly expressed in the heart, and known to be upregulated in HF. Over the last 20 years, both GRK2 and GRK5 have been demonstrated to be critical mediators of the molecular alterations that occur in the failing heart. In the present review, we will highlight recent findings that further characterize "non-canonical" GRK signaling observed in HF. Further, we will also present potential therapeutic strategies (i.e. small molecule inhibition, microRNAs, gene therapy) that may have potential in combating the deleterious effects of GRKs in HF.
Collapse
Affiliation(s)
| | - Christopher J Traynham
- Center for Translational Medicine, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, United States
| | - Ryan C Coleman
- Center for Translational Medicine, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, United States
| | - Walter J Koch
- Center for Translational Medicine, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, United States.
| |
Collapse
|
26
|
Penela P. Chapter Three - Ubiquitination and Protein Turnover of G-Protein-Coupled Receptor Kinases in GPCR Signaling and Cellular Regulation. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 141:85-140. [PMID: 27378756 DOI: 10.1016/bs.pmbts.2016.04.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
G-protein-coupled receptors (GPCRs) are responsible for regulating a wide variety of physiological processes, and distinct mechanisms for GPCR inactivation exist to guarantee correct receptor functionality. One of the widely used mechanisms is receptor phosphorylation by specific G-protein-coupled receptor kinases (GRKs), leading to uncoupling from G proteins (desensitization) and receptor internalization. GRKs and β-arrestins also participate in the assembly of receptor-associated multimolecular complexes, thus initiating alternative G-protein-independent signaling events. In addition, the abundant GRK2 kinase has diverse "effector" functions in cellular migration, proliferation, and metabolism homeostasis by means of the phosphorylation or interaction with non-GPCR partners. Altered expression of GRKs (particularly of GRK2 and GRK5) occurs during pathological conditions characterized by impaired GPCR signaling including inflammatory syndromes, cardiovascular disease, and tumor contexts. It is increasingly appreciated that different pathways governing GRK protein stability play a role in the modulation of kinase levels in normal and pathological conditions. Thus, enhanced GRK2 degradation by the proteasome pathway occurs upon GPCR stimulation, what allows cellular adaptation to chronic stimulation in a physiological setting. β-arrestins participate in this process by facilitating GRK2 phosphorylation by different kinases and by recruiting diverse E3 ubiquitin ligase to the receptor complex. Different proteolytic systems (ubiquitin-proteasome, calpains), chaperone activities and signaling pathways influence the stability of GRKs in different ways, thus endowing specificity to GPCR regulation as protein turnover of GRKs can be differentially affected. Therefore, modulation of protein stability of GRKs emerges as a versatile mechanism for feedback regulation of GPCR signaling and basic cellular processes.
Collapse
Affiliation(s)
- P Penela
- Department of Molecular Biology and Centre of Molecular Biology "Severo Ochoa" (CSIC-UAM), Madrid, Autonomous University of Madrid, Madrid, Spain; Spain Health Research Institute The Princesa, Madrid, Spain.
| |
Collapse
|
27
|
Yang P, Homan KT, Li Y, Cruz-Rodríguez O, Tesmer JJG, Chen Z. Effect of Lipid Composition on the Membrane Orientation of the G Protein-Coupled Receptor Kinase 2-Gβ1γ2 Complex. Biochemistry 2016; 55:2841-8. [PMID: 27088923 DOI: 10.1021/acs.biochem.6b00354] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Interactions between proteins and cell membranes are critical for biological processes such as transmembrane signaling, and specific components of the membrane may play roles in helping to organize or mandate particular conformations of both integral and peripheral membrane proteins. One example of a signaling enzyme whose function is dependent on membrane binding and whose activity is affected by specific lipid components is G protein-coupled receptor (GPCR) kinase 2 (GRK2). Efficient GRK2-mediated phosphorylation of activated GPCRs is dependent not only on its recruitment to the membrane by heterotrimeric Gβγ subunits but also on the presence of highly negatively charged lipids, in particular phosphatidylinositol 4',5'-bisphosphate (PIP2). We hypothesized that PIP2 may favor a distinct orientation of the GRK2-Gβγ complex on the membrane that is more optimal for function. In this study, we compared the possible orientations of the GRK2-Gβγ complex and Gβγ alone on model cell membranes prepared with various anionic phospholipids as deduced from sum frequency generation vibrational and attenuated total reflectance Fourier transform infrared spectroscopic methods. Our results indicate that PIP2 affects the membrane orientation of the GRK2-Gβ1γ2 complex but not that of complexes formed with anionic phospholipid binding deficient mutations in the GRK2 pleckstrin homology (PH) domain. Gβ1γ2 exhibits a similar orientation on the lipid bilayer regardless of its lipid composition. The PIP2-induced orientation of the GRK2-Gβ1γ2 complex is therefore most likely caused by specific interactions between PIP2 and the GRK2 PH domain. Thus, PIP2 not only helps recruit GRK2 to the membrane but also "fine tunes" the orientation of the GRK2-Gβγ complex so that it is better positioned to phosphorylate activated GPCRs.
Collapse
Affiliation(s)
- Pei Yang
- Department of Chemistry, University of Michigan , 930 North University Avenue, Ann Arbor, Michigan 48109, United States
| | - Kristoff T Homan
- Departments of Pharmacology and Biological Chemistry, Life Sciences Institute, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Yaoxin Li
- Department of Chemistry, University of Michigan , 930 North University Avenue, Ann Arbor, Michigan 48109, United States
| | - Osvaldo Cruz-Rodríguez
- Departments of Pharmacology and Biological Chemistry, Life Sciences Institute, University of Michigan , Ann Arbor, Michigan 48109, United States.,Ph.D. Program in Chemical Biology, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - John J G Tesmer
- Departments of Pharmacology and Biological Chemistry, Life Sciences Institute, University of Michigan , Ann Arbor, Michigan 48109, United States
| | - Zhan Chen
- Department of Chemistry, University of Michigan , 930 North University Avenue, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
28
|
Traynham CJ, Hullmann J, Koch WJ. "Canonical and non-canonical actions of GRK5 in the heart". J Mol Cell Cardiol 2016; 92:196-202. [PMID: 26829117 PMCID: PMC4789097 DOI: 10.1016/j.yjmcc.2016.01.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 01/13/2016] [Accepted: 01/28/2016] [Indexed: 01/13/2023]
Abstract
As the average world-wide lifespan continues to increase, heart failure (HF) has dramatically increased in incidence leading to the highest degree of mortality and morbidity of any disease presently studied. G protein-coupled receptors (GPCRs) play a prominent role in regulation of cardiovascular function. GPCRs are effectively "turned off" by GPCR kinases (GRKs) in a process known as "desensitization". GRKs 2 and 5 are highly expressed in the heart, and known to be upregulated in HF. Over the last 20years, the role of GRK2 in HF has been widely studied. However, until recently, the role of GRK5 in cardiac pathophysiology had yet to be elucidated. In the present review, we will focus on GRK5's role in the myocardium in normal physiology, and its apparent critical role in the progression of HF. Further, we will also present potential therapeutic strategies (i.e. small molecule inhibition, gene therapy) that may have potential in combating the deleterious effects of GRK5 in HF.
Collapse
Affiliation(s)
- Christopher J Traynham
- Center for Translational Medicine, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, United States
| | | | - Walter J Koch
- Center for Translational Medicine, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA 19140, United States.
| |
Collapse
|
29
|
“Barcode” and Differential Effects of GPCR Phosphorylation by Different GRKs. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2016. [DOI: 10.1007/978-1-4939-3798-1_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
30
|
Zheng M, Zhang X, Guo S, Zhang X, Min C, Cheon SH, Oak MH, Kim YR, Kim KM. Agonist-induced changes in RalA activities allows the prediction of the endocytosis of G protein-coupled receptors. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:77-90. [PMID: 26477566 DOI: 10.1016/j.bbamcr.2015.10.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 10/10/2015] [Accepted: 10/13/2015] [Indexed: 12/31/2022]
Abstract
GTP binding proteins are classified into two families: heterotrimeric large G proteins which are composed of three subunits, and one subunit of small G proteins. Roles of small G proteins in the intracellular trafficking of G protein-coupled receptors (GPCRs) were studied. Among various small G proteins tested, GTP-bound form (G23V) of RalA inhibited the internalization of dopamine D2 receptor independently of the previously reported downstream effectors of RalA, such as Ral-binding protein 1 and PLD. With high affinity for GRK2, active RalA inhibited the GPCR endocytosis by sequestering the GRK2 from receptors. When it was tested for several GPCRs including an endogenous GPCR, lysophosphatidic acid receptor 1, agonist-induced conversion of GTP-bound to GDP-bound RalA, which presumably releases the sequestered GRK2, was observed selectively with the GPCRs which have tendency to undergo endocytosis. Conversion of RalA from active to inactive state occurred by translocation of RGL, a guanine nucleotide exchange factor, from the plasma membrane to cytosol as a complex with Gβγ. These results suggest that agonist-induced Gβγ-mediated conversion of RalA from the GTP-bound form to the GDP-bound form could be a mechanism to facilitate agonist-induced internalization of GPCRs.
Collapse
Affiliation(s)
- Mei Zheng
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Xiaohan Zhang
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Shuohan Guo
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Xiaowei Zhang
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Chengchun Min
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Seung Hoon Cheon
- Department of Medicinal Chemistry, College of Pharmacy, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Min-Ho Oak
- College of Pharmacy, Mokpo National University, Muan-gun, Jeollanamdo 534-729, Republic of Korea
| | - Young Ran Kim
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 500-757, Republic of Korea
| | - Kyeong-Man Kim
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 500-757, Republic of Korea.
| |
Collapse
|
31
|
Abstract
G-protein-coupled receptor kinases (GRKs) are serine/threonine protein kinases originally discovered for their role in G-protein-coupled receptor (GPCR) phosphorylation. Recent studies have demonstrated a much broader function for this kinase family including phosphorylation of cytosolic substrates involved in cell signaling pathways stimulated by GPCRs, as well as by non-GPCRs. In addition, GRKs modulate signaling via phosphorylation-independent functions. Because of these various biochemical functions, GRKs have been shown to affect critical physiological and pathophysiological processes, and thus are considered as drug targets in diseases such as heart failure. Role of GRKs in inflammation and inflammatory diseases is an evolving area of research and several studies including work from our lab in the recent years have demonstrated critical role of GRKs in the immune system. In this review, we discuss the classical and the newly emerging functions of GRKs in the immune system and their role in inflammation and disease processes.
Collapse
|
32
|
Inagaki S, Ghirlando R, Vishnivetskiy S, Homan KT, White JF, Tesmer JJG, Gurevich VV, Grisshammer R. G Protein-Coupled Receptor Kinase 2 (GRK2) and 5 (GRK5) Exhibit Selective Phosphorylation of the Neurotensin Receptor in Vitro. Biochemistry 2015; 54:4320-4329. [PMID: 26120872 PMCID: PMC4512254 DOI: 10.1021/acs.biochem.5b00285] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 06/05/2015] [Indexed: 12/24/2022]
Abstract
G protein-coupled receptor kinases (GRKs) play an important role in the desensitization of G protein-mediated signaling of G protein-coupled receptors (GPCRs). The level of interest in mapping their phosphorylation sites has increased because recent studies suggest that the differential pattern of receptor phosphorylation has distinct biological consequences. In vitro phosphorylation experiments using well-controlled systems are useful for deciphering the complexity of these physiological reactions and understanding the targeted event. Here, we report on the phosphorylation of the class A GPCR neurotensin receptor 1 (NTSR1) by GRKs under defined experimental conditions afforded by nanodisc technology. Phosphorylation of NTSR1 by GRK2 was agonist-dependent, whereas phosphorylation by GRK5 occurred in an activation-independent manner. In addition, the negatively charged lipids in the immediate vicinity of NTSR1 directly affect phosphorylation by GRKs. Identification of phosphorylation sites in agonist-activated NTSR1 revealed that GRK2 and GRK5 target different residues located on the intracellular receptor elements. GRK2 phosphorylates only the C-terminal Ser residues, whereas GRK5 phosphorylates Ser and Thr residues located in intracellular loop 3 and the C-terminus. Interestingly, phosphorylation assays using a series of NTSR1 mutants show that GRK2 does not require acidic residues upstream of the phospho-acceptors for site-specific phosphorylation, in contrast to the β2-adrenergic and μ-opioid receptors. Differential phosphorylation of GPCRs by GRKs is thought to encode a particular signaling outcome, and our in vitro study revealed NTSR1 differential phosphorylation by GRK2 and GRK5.
Collapse
Affiliation(s)
- Sayaka Inagaki
- Membrane
Protein Structure Function Unit, National Institute of Neurological
Disorders and Stroke, National Institutes
of Health, Department of Health and Human Services, Rockville, Maryland 20852, United States
| | - Rodolfo Ghirlando
- Laboratory
of Molecular Biology, National Institute of Diabetes and Digestive
and Kidney Diseases, National Institutes
of Health, Department of Health and Human Services, Bethesda, Maryland 20892, United States
| | - Sergey
A. Vishnivetskiy
- Department
of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Kristoff T. Homan
- Departments
of Pharmacology and Biological Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jim F. White
- Membrane
Protein Structure Function Unit, National Institute of Neurological
Disorders and Stroke, National Institutes
of Health, Department of Health and Human Services, Rockville, Maryland 20852, United States
| | - John J. G. Tesmer
- Departments
of Pharmacology and Biological Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Vsevolod V. Gurevich
- Department
of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Reinhard Grisshammer
- Membrane
Protein Structure Function Unit, National Institute of Neurological
Disorders and Stroke, National Institutes
of Health, Department of Health and Human Services, Rockville, Maryland 20852, United States
| |
Collapse
|
33
|
Allen SJ, Parthasarathy G, Darke PL, Diehl RE, Ford RE, Hall DL, Johnson SA, Reid JC, Rickert KW, Shipman JM, Soisson SM, Zuck P, Munshi SK, Lumb KJ. Structure and Function of the Hypertension Variant A486V of G Protein-coupled Receptor Kinase 4. J Biol Chem 2015; 290:20360-73. [PMID: 26134571 DOI: 10.1074/jbc.m115.648907] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Indexed: 11/06/2022] Open
Abstract
G-protein-coupled receptor (GPCR) kinases (GRKs) bind to and phosphorylate GPCRs, initiating the process of GPCR desensitization and internalization. GRK4 is implicated in the regulation of blood pressure, and three GRK4 polymorphisms (R65L, A142V, and A486V) are associated with hypertension. Here, we describe the 2.6 Å structure of human GRK4α A486V crystallized in the presence of 5'-adenylyl β,γ-imidodiphosphate. The structure of GRK4α is similar to other GRKs, although slight differences exist within the RGS homology (RH) bundle subdomain, substrate-binding site, and kinase C-tail. The RH bundle subdomain and kinase C-terminal lobe form a strikingly acidic surface, whereas the kinase N-terminal lobe and RH terminal subdomain surfaces are much more basic. In this respect, GRK4α is more similar to GRK2 than GRK6. A fully ordered kinase C-tail reveals interactions linking the C-tail with important determinants of kinase activity, including the αB helix, αD helix, and the P-loop. Autophosphorylation of wild-type GRK4α is required for full kinase activity, as indicated by a lag in phosphorylation of a peptide from the dopamine D1 receptor without ATP preincubation. In contrast, this lag is not observed in GRK4α A486V. Phosphopeptide mapping by mass spectrometry indicates an increased rate of autophosphorylation of a number of residues in GRK4α A486V relative to wild-type GRK4α, including Ser-485 in the kinase C-tail.
Collapse
Affiliation(s)
- Samantha J Allen
- From Screening and Protein Sciences, Merck Research Laboratories, North Wales, Pennsylvania 19454 and
| | - Gopal Parthasarathy
- Structural Chemistry, Merck Research Laboratories, West Point, Pennsylvania 19486
| | - Paul L Darke
- From Screening and Protein Sciences, Merck Research Laboratories, North Wales, Pennsylvania 19454 and
| | - Ronald E Diehl
- From Screening and Protein Sciences, Merck Research Laboratories, North Wales, Pennsylvania 19454 and
| | - Rachael E Ford
- From Screening and Protein Sciences, Merck Research Laboratories, North Wales, Pennsylvania 19454 and
| | - Dawn L Hall
- From Screening and Protein Sciences, Merck Research Laboratories, North Wales, Pennsylvania 19454 and
| | - Scott A Johnson
- Structural Chemistry, Merck Research Laboratories, West Point, Pennsylvania 19486
| | - John C Reid
- Structural Chemistry, Merck Research Laboratories, West Point, Pennsylvania 19486
| | - Keith W Rickert
- From Screening and Protein Sciences, Merck Research Laboratories, North Wales, Pennsylvania 19454 and
| | - Jennifer M Shipman
- From Screening and Protein Sciences, Merck Research Laboratories, North Wales, Pennsylvania 19454 and
| | - Stephen M Soisson
- Structural Chemistry, Merck Research Laboratories, West Point, Pennsylvania 19486
| | - Paul Zuck
- From Screening and Protein Sciences, Merck Research Laboratories, North Wales, Pennsylvania 19454 and
| | - Sanjeev K Munshi
- From Screening and Protein Sciences, Merck Research Laboratories, North Wales, Pennsylvania 19454 and
| | - Kevin J Lumb
- From Screening and Protein Sciences, Merck Research Laboratories, North Wales, Pennsylvania 19454 and
| |
Collapse
|
34
|
Komolov KE, Bhardwaj A, Benovic JL. Atomic Structure of GRK5 Reveals Distinct Structural Features Novel for G Protein-coupled Receptor Kinases. J Biol Chem 2015; 290:20629-20647. [PMID: 26032409 DOI: 10.1074/jbc.m115.647297] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Indexed: 11/06/2022] Open
Abstract
G protein-coupled receptor kinases (GRKs) are members of the protein kinase A, G, and C families (AGC) and play a central role in mediating G protein-coupled receptor phosphorylation and desensitization. One member of the family, GRK5, has been implicated in several human pathologies, including heart failure, hypertension, cancer, diabetes, and Alzheimer disease. To gain mechanistic insight into GRK5 function, we determined a crystal structure of full-length human GRK5 at 1.8 Å resolution. GRK5 in complex with the ATP analog 5'-adenylyl β,γ-imidodiphosphate or the nucleoside sangivamycin crystallized as a monomer. The C-terminal tail (C-tail) of AGC kinase domains is a highly conserved feature that is divided into three segments as follows: the C-lobe tether, the active-site tether (AST), and the N-lobe tether (NLT). This domain is fully resolved in GRK5 and reveals novel interactions with the nucleotide and N-lobe. Similar to other AGC kinases, the GRK5 AST is an integral part of the nucleotide-binding pocket, a feature not observed in other GRKs. The AST also mediates contact between the kinase N- and C-lobes facilitating closure of the kinase domain. The GRK5 NLT is largely displaced from its previously observed position in other GRKs. Moreover, although the autophosphorylation sites in the NLT are >20 Å away from the catalytic cleft, they are capable of rapid cis-autophosphorylation suggesting high mobility of this region. In summary, we provide a snapshot of GRK5 in a partially closed state, where structural elements of the kinase domain C-tail are aligned to form novel interactions to the nucleotide and N-lobe not previously observed in other GRKs.
Collapse
Affiliation(s)
- Konstantin E Komolov
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Anshul Bhardwaj
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107.
| | - Jeffrey L Benovic
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107.
| |
Collapse
|
35
|
Homan KT, Waldschmidt HV, Glukhova A, Cannavo A, Song J, Cheung JY, Koch WJ, Larsen SD, Tesmer JJG. Crystal Structure of G Protein-coupled Receptor Kinase 5 in Complex with a Rationally Designed Inhibitor. J Biol Chem 2015; 290:20649-20659. [PMID: 26032411 DOI: 10.1074/jbc.m115.647370] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Indexed: 11/06/2022] Open
Abstract
G protein-coupled receptor kinases (GRKs) regulate cell signaling by initiating the desensitization of active G protein-coupled receptors. The two most widely expressed GRKs (GRK2 and GRK5) play a role in cardiovascular disease and thus represent important targets for the development of novel therapeutic drugs. In the course of a GRK2 structure-based drug design campaign, one inhibitor (CCG215022) exhibited nanomolar IC50 values against both GRK2 and GRK5 and good selectivity against other closely related kinases such as GRK1 and PKA. Treatment of murine cardiomyocytes with CCG215022 resulted in significantly increased contractility at 20-fold lower concentrations than paroxetine, an inhibitor with more modest selectivity for GRK2. A 2.4 Å crystal structure of the GRK5·CCG215022 complex was determined and revealed that the inhibitor binds in the active site similarly to its parent compound GSK180736A. As designed, its 2-pyridylmethyl amide side chain occupies the hydrophobic subsite of the active site where it forms three additional hydrogen bonds, including one with the catalytic lysine. The overall conformation of the GRK5 kinase domain is similar to that of a previously determined structure of GRK6 in what is proposed to be its active state, but the C-terminal region of the enzyme adopts a distinct conformation. The kinetic properties of site-directed mutants in this region are consistent with the hypothesis that this novel C-terminal structure is representative of the membrane-bound conformation of the enzyme.
Collapse
Affiliation(s)
- Kristoff T Homan
- Life Sciences Institute and the Departments of Pharmacology and Biological Sciences, University of Michigan, Ann Arbor, Michigan 48109
| | - Helen V Waldschmidt
- Vahlteich Medicinal Chemistry Core and the Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Alisa Glukhova
- Life Sciences Institute and the Departments of Pharmacology and Biological Sciences, University of Michigan, Ann Arbor, Michigan 48109
| | - Alessandro Cannavo
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Jianliang Song
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Joseph Y Cheung
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Walter J Koch
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| | - Scott D Larsen
- Vahlteich Medicinal Chemistry Core and the Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - John J G Tesmer
- Life Sciences Institute and the Departments of Pharmacology and Biological Sciences, University of Michigan, Ann Arbor, Michigan 48109.
| |
Collapse
|
36
|
Sato PY, Chuprun JK, Schwartz M, Koch WJ. The evolving impact of g protein-coupled receptor kinases in cardiac health and disease. Physiol Rev 2015; 95:377-404. [PMID: 25834229 PMCID: PMC4551214 DOI: 10.1152/physrev.00015.2014] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are important regulators of various cellular functions via activation of intracellular signaling events. Active GPCR signaling is shut down by GPCR kinases (GRKs) and subsequent β-arrestin-mediated mechanisms including phosphorylation, internalization, and either receptor degradation or resensitization. The seven-member GRK family varies in their structural composition, cellular localization, function, and mechanism of action (see sect. II). Here, we focus our attention on GRKs in particular canonical and novel roles of the GRKs found in the cardiovascular system (see sects. III and IV). Paramount to overall cardiac function is GPCR-mediated signaling provided by the adrenergic system. Overstimulation of the adrenergic system has been highly implicated in various etiologies of cardiovascular disease including hypertension and heart failure. GRKs acting downstream of heightened adrenergic signaling appear to be key players in cardiac homeostasis and disease progression, and herein we review the current data on GRKs related to cardiac disease and discuss their potential in the development of novel therapeutic strategies in cardiac diseases including heart failure.
Collapse
Affiliation(s)
- Priscila Y Sato
- Center for Translational Medicine and Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania; and Advanced Institutes of Convergence Technology, Suwon, South Korea
| | - J Kurt Chuprun
- Center for Translational Medicine and Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania; and Advanced Institutes of Convergence Technology, Suwon, South Korea
| | - Mathew Schwartz
- Center for Translational Medicine and Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania; and Advanced Institutes of Convergence Technology, Suwon, South Korea
| | - Walter J Koch
- Center for Translational Medicine and Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania; and Advanced Institutes of Convergence Technology, Suwon, South Korea
| |
Collapse
|
37
|
Hullmann JE, Grisanti LA, Makarewich CA, Gao E, Gold JI, Chuprun JK, Tilley DG, Houser SR, Koch WJ. GRK5-mediated exacerbation of pathological cardiac hypertrophy involves facilitation of nuclear NFAT activity. Circ Res 2014; 115:976-85. [PMID: 25332207 PMCID: PMC4258119 DOI: 10.1161/circresaha.116.304475] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 10/20/2014] [Indexed: 01/19/2023]
Abstract
RATIONALE G protein-coupled receptor kinases (GRKs) acting in the cardiomyocyte regulate important signaling events that control cardiac function. Both GRK2 and GRK5, the predominant GRKs expressed in the heart, have been shown to be upregulated in failing human myocardium. Although the canonical role of GRKs is to desensitize G protein-coupled receptors via phosphorylation, it has been demonstrated that GRK5, unlike GRK2, can reside in the nucleus of myocytes and exert G protein-coupled receptor-independent effects that promote maladaptive cardiac hypertrophy and heart failure. OBJECTIVE To explore novel mechanisms by which GRK5 acting in the nucleus of cardiomyocytes participates in pathological cardiac hypertrophy. METHODS AND RESULTS In this study, we have found that GRK5-mediated pathological cardiac hypertrophy involves the activation of the nuclear factor of activated T cells (NFAT) because GRK5 causes enhancement of NFAT-mediated hypertrophic gene transcription. Transgenic mice with cardiomyocyte-specific GRK5 overexpression activate an NFAT-reporter in mice basally and after hypertrophic stimulation, including transverse aortic constriction and phenylephrine treatment. Complimentary to this, GRK5 null mice exhibit less NFAT transcriptional activity after transverse aortic constriction. Furthermore, the loss of NFATc3 expression in the heart protected GRK5 overexpressing transgenic mice from the exaggerated hypertrophy and early progression to heart failure seen after transverse aortic constriction. Molecular studies suggest that GRK5 acts in concert with NFAT to increase hypertrophic gene transcription in the nucleus via GRK5's ability to bind DNA directly without a phosphorylation event. CONCLUSIONS GRK5, acting in a kinase independent manner, is a facilitator of NFAT activity and part of a DNA-binding complex responsible for pathological hypertrophic gene transcription.
Collapse
Affiliation(s)
- Jonathan E Hullmann
- From the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA (J.E.H., J.I.G.); and Center for Translational Medicine (J.E.H., L.A.G., E.G. J.I.G., J.K.C., D.G.T., W.J.K.) and Cardiovascular Research Center (C.A.M., S.R.H.), Temple University School of Medicine, Philadelphia, PA
| | - Laurel A Grisanti
- From the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA (J.E.H., J.I.G.); and Center for Translational Medicine (J.E.H., L.A.G., E.G. J.I.G., J.K.C., D.G.T., W.J.K.) and Cardiovascular Research Center (C.A.M., S.R.H.), Temple University School of Medicine, Philadelphia, PA
| | - Catherine A Makarewich
- From the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA (J.E.H., J.I.G.); and Center for Translational Medicine (J.E.H., L.A.G., E.G. J.I.G., J.K.C., D.G.T., W.J.K.) and Cardiovascular Research Center (C.A.M., S.R.H.), Temple University School of Medicine, Philadelphia, PA
| | - Erhe Gao
- From the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA (J.E.H., J.I.G.); and Center for Translational Medicine (J.E.H., L.A.G., E.G. J.I.G., J.K.C., D.G.T., W.J.K.) and Cardiovascular Research Center (C.A.M., S.R.H.), Temple University School of Medicine, Philadelphia, PA
| | - Jessica I Gold
- From the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA (J.E.H., J.I.G.); and Center for Translational Medicine (J.E.H., L.A.G., E.G. J.I.G., J.K.C., D.G.T., W.J.K.) and Cardiovascular Research Center (C.A.M., S.R.H.), Temple University School of Medicine, Philadelphia, PA
| | - J Kurt Chuprun
- From the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA (J.E.H., J.I.G.); and Center for Translational Medicine (J.E.H., L.A.G., E.G. J.I.G., J.K.C., D.G.T., W.J.K.) and Cardiovascular Research Center (C.A.M., S.R.H.), Temple University School of Medicine, Philadelphia, PA
| | - Douglas G Tilley
- From the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA (J.E.H., J.I.G.); and Center for Translational Medicine (J.E.H., L.A.G., E.G. J.I.G., J.K.C., D.G.T., W.J.K.) and Cardiovascular Research Center (C.A.M., S.R.H.), Temple University School of Medicine, Philadelphia, PA
| | - Steven R Houser
- From the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA (J.E.H., J.I.G.); and Center for Translational Medicine (J.E.H., L.A.G., E.G. J.I.G., J.K.C., D.G.T., W.J.K.) and Cardiovascular Research Center (C.A.M., S.R.H.), Temple University School of Medicine, Philadelphia, PA
| | - Walter J Koch
- From the Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA (J.E.H., J.I.G.); and Center for Translational Medicine (J.E.H., L.A.G., E.G. J.I.G., J.K.C., D.G.T., W.J.K.) and Cardiovascular Research Center (C.A.M., S.R.H.), Temple University School of Medicine, Philadelphia, PA.
| |
Collapse
|
38
|
Santiago FE, Fior-Chadi DR, Carrettiero DC. Alpha2-adrenoceptor and adenosine A1 receptor within the nucleus tractus solitarii in hypertension development. Auton Neurosci 2014; 187:36-44. [PMID: 25466830 DOI: 10.1016/j.autneu.2014.11.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 10/21/2014] [Accepted: 11/01/2014] [Indexed: 02/07/2023]
Abstract
Alpha2-adrenoceptor and A1 adenosine receptor systems within the nucleus tractus solitarii (NTS) play an important role in cardiovascular control. Deregulation of these systems may result in an elevated sympathetic tone, one of the root causes of neurogenic hypertension. The dorsomedial/dorsolateral and subpostremal NTS subnuclei of spontaneously hypertensive rats (SHR) show density changes in both receptors, even at 15 days of age, prior to the onset of hypertension. In addition, adenosine A1 receptors have been specifically reported to modulate alpha2-adrenoceptors in several brain regions, including the NTS, via a PLC-dependent pathway involving cross regulation between sympathetic neurons and astrocytes. The physiological cross talk between these receptor systems is also deregulated in SHR suggesting that alpha2-adrenoceptor and A1 adenosine receptor might be germane to the development of hypertension. In this review, we will focus on these systems within the NTS during development, pointing out some interesting modulations in processes, and chemical changes within specific subnuclei of NTS circuitry, that might have implications for neurogenic hypertension.
Collapse
Affiliation(s)
- Fernando E Santiago
- Universidade Federal do ABC (UFABC), Centro de Ciências Naturais e Humanas, Santo André, SP, Brazil
| | - Débora R Fior-Chadi
- Universidade de São Paulo (USP), Departamento de Fisiologia, Instituto de Biociências, São Paulo, SP, Brazil
| | - Daniel C Carrettiero
- Universidade Federal do ABC (UFABC), Centro de Ciências Naturais e Humanas, Santo André, SP, Brazil.
| |
Collapse
|
39
|
Xu H, Jiang X, Shen K, Fischer CC, Wedegaertner PB. The regulator of G protein signaling (RGS) domain of G protein-coupled receptor kinase 5 (GRK5) regulates plasma membrane localization and function. Mol Biol Cell 2014; 25:2105-15. [PMID: 24807909 PMCID: PMC4072583 DOI: 10.1091/mbc.e13-09-0547] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
GRK5/GRK4 chimeras and point mutations in GRK5 identify a short sequence within the RGS domain in GRK5 that is critical for GRK5 PM localization. CoIP and acceptor photobleaching FRET assays show that expressed GRK5 self-associates in cells, and RGS domain mutations disrupt the coIP. Taken together, the results provide evidence for a novel mechanism in which RGS domain–mediated dimerization is necessary for plasma membrane localization of GRK5. The G protein–coupled receptor (GPCR) kinases (GRKs) phosphorylate activated GPCRs at the plasma membrane (PM). Here GRK5/GRK4 chimeras and point mutations in GRK5 identify a short sequence within the regulator of G protein signaling (RGS) domain in GRK5 that is critical for GRK5 PM localization. This region of the RGS domain of GRK5 coincides with a region of GRK6 and GRK1 shown to form a hydrophobic dimeric interface (HDI) in crystal structures. Coimmunoprecipitation (coIP) and acceptor photobleaching fluorescence resonance energy transfer assays show that expressed GRK5 self-associates in cells, whereas GRK5-M165E/F166E (GRK5-EE), containing hydrophilic mutations in the HDI region of the RGS domain, displays greatly decreased coIP interactions. Both forcing dimerization of GRK5-EE, via fusion to leucine zipper motifs, and appending an extra C-terminal membrane-binding region to GRK5-EE (GRK5-EE-CT) recover PM localization. In addition, GRK5-EE displays a decreased ability to inhibit PAR1-induced calcium release compared with GRK5 wild type (wt). In contrast, PM-localized GRK5-EE-CaaX (appending a C-terminal prenylation and polybasic motif from K-ras) or GRK5-EE-CT shows comparable ability to GRK5 wt to inhibit PAR1-induced calcium release. The results suggest a novel model in which GRK5 dimerization is important for its plasma membrane localization and function.
Collapse
Affiliation(s)
- Hua Xu
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Xiaoshan Jiang
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107Center for Science Research, Guilin Medical University, Guilin, Guangxi 541004, China
| | - Ke Shen
- Center for Science Research, Guilin Medical University, Guilin, Guangxi 541004, China
| | - Christopher C Fischer
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107
| | - Philip B Wedegaertner
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
40
|
Ding B, Glukhova A, Sobczyk-Kojiro K, Mosberg HI, Tesmer JJG, Chen Z. Unveiling the membrane-binding properties of N-terminal and C-terminal regions of G protein-coupled receptor kinase 5 by combined optical spectroscopies. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2014; 30:823-31. [PMID: 24401145 PMCID: PMC3983354 DOI: 10.1021/la404055a] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 12/21/2013] [Indexed: 06/03/2023]
Abstract
G protein-coupled receptor kinase 5 (GRK5) is thought to associate with membranes in part via N- and C-terminal segments that are typically disordered in available high-resolution crystal structures. Herein we investigate the interactions of these regions with model cell membrane using combined sum frequency generation (SFG) vibrational spectroscopy and attenuated total reflectance-Fourier transform infrared (ATR-FTIR) spectroscopy. It was found that both regions associate with POPC lipid bilayers but adopt different structures when doing so: GRK5 residues 2-31 (GRK5(2-31)) was in random coil whereas GRK5(546-565) was partially helical. When the subphase for the GRK5(2-31) peptide was changed to 40% TFE/60% 10 mM phosphate pH 7.4 buffer, a large change in the SFG amide I signal indicated that GRK5(2-31) became partially helical. By inspecting the membrane behavior of two different segments of GRK5(2-31), namely, GRK5(2-24) and GRK5(25-31), we found that residues 25-31 are responsible for membrane binding, whereas the helical character is imparted by residues 2-24. With SFG, we deduced that the orientation angle of the helical segment of GRK5(2-31) is 46 ± 1° relative to the surface normal in 40% TFE/60% 10 mM phosphate pH = 7.4 buffer but increases to 78 ± 11° with higher ionic strength. We also investigated the effect of PIP2 in the model membrane and concluded that the POPC:PIP2 (9:1) lipid bilayer did not change the behavior of either peptide compared to a pure POPC lipid bilayer. With ATR-FTIR, we also found that Ca(2+)·calmodulin is able to extract both peptides from the POPC lipid bilayer, consistent with the role of this protein in disrupting GRK5 interactions with the plasma membrane in cells.
Collapse
Affiliation(s)
- Bei Ding
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Alisa Glukhova
- Life
Sciences Institute and the Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109-2216, United States
| | - Katarzyna Sobczyk-Kojiro
- College
of Pharmacy, Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109-1065, United States
| | - Henry I. Mosberg
- College
of Pharmacy, Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan 48109-1065, United States
| | - John J. G. Tesmer
- Life
Sciences Institute and the Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109-2216, United States
| | - Zhan Chen
- Department
of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
41
|
Yang P, Glukhova A, Tesmer JJG, Chen Z. Membrane orientation and binding determinants of G protein-coupled receptor kinase 5 as assessed by combined vibrational spectroscopic studies. PLoS One 2013; 8:e82072. [PMID: 24278472 PMCID: PMC3838385 DOI: 10.1371/journal.pone.0082072] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Accepted: 10/20/2013] [Indexed: 11/18/2022] Open
Abstract
G-protein coupled receptors (GPCRs) are integral membrane proteins involved in a wide variety of biological processes in eukaryotic cells, and are targeted by a large fraction of marketed drugs. GPCR kinases (GRKs) play important roles in feedback regulation of GPCRs, such as of β-adrenergic receptors in the heart, where GRK2 and GRK5 are the major isoforms expressed. Membrane targeting is essential for GRK function in cells. Whereas GRK2 is recruited to the membrane by heterotrimeric Gβγ subunits, the mechanism of membrane binding by GRK5 is not fully understood. It has been proposed that GRK5 is constitutively associated with membranes through elements located at its N-terminus, its C-terminus, or both. The membrane orientation of GRK5 is also a matter of speculation. In this work, we combined sum frequency generation (SFG) vibrational spectroscopy and attenuated total reflectance-Fourier transform infrared spectroscopy (ATR-FTIR) to help determine the membrane orientation of GRK5 and a C-terminally truncated mutant (GRK51-531) on membrane lipid bilayers. It was found that GRK5 and GRK51-531 adopt a similar orientation on model cell membranes in the presence of PIP2 that is similar to that predicted for GRK2 in prior studies. Mutation of the N-terminal membrane binding site of GRK5 did not eliminate membrane binding, but prevented observation of this discrete orientation. The C-terminus of GRK5 does not have substantial impact on either membrane binding or orientation in this model system. Thus, the C-terminus of GRK5 may drive membrane binding in cells via interactions with other proteins at the plasma membrane or bind in an unstructured manner to negatively charged membranes.
Collapse
Affiliation(s)
- Pei Yang
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Alisa Glukhova
- Departments of Pharmacology and Biological Chemistry, Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- Program in Chemical Biology, University of Michigan, Ann Arbor, Michigan, United States of America
| | - John J. G. Tesmer
- Departments of Pharmacology and Biological Chemistry, Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail: (ZC); (JJGT)
| | - Zhan Chen
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail: (ZC); (JJGT)
| |
Collapse
|
42
|
Johnson LR, Robinson JD, Lester KN, Pitcher JA. Distinct structural features of G protein-coupled receptor kinase 5 (GRK5) regulate its nuclear localization and DNA-binding ability. PLoS One 2013; 8:e62508. [PMID: 23658733 PMCID: PMC3642199 DOI: 10.1371/journal.pone.0062508] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 03/26/2013] [Indexed: 12/26/2022] Open
Abstract
G protein-coupled receptor kinases (GRKs) act to desensitize G protein-coupled receptors (GPCRs). In addition to this role at the plasma membrane, a nuclear function for GRK5, a member of the GRK4 subfamily of GRKs, has been reported. GRK5 phosphorylates and promotes the nuclear export of the histone deacetylase, HDAC5. Here we demonstrate that the possession of a nuclear localization sequence (NLS) is a common feature of GRK4 subfamily members (GRKs 4, 5 and 6). However, the location of the NLS and the ability of these GRKs to bind DNA in vitro are different. The NLSs of GRK5 and 6 bind DNA in vitro, whilst the NLS of GRK4 does not. Using mutants of GRK5 we identify the regions of GRK5 required for DNA-binding in vitro and nuclear localization in cells. The DNA-binding ability of GRK5 requires both the NLS and an N-terminal calmodulin (CaM)-binding site. A functional nuclear export sequence (NES), required for CaM-dependent nuclear export of the kinase, is also identified. Based on our observations we propose a model to explain how nuclear localization of GRK5 may be regulated. Notably, the nuclear localization of GRK5 and 6 is differentially regulated. These results suggest subfamily specific nuclear functions for the GRK4 subfamily members. Identification of GRK specific small molecule inhibitors of nuclear localization and/or function for the GRK4 subfamily may thus be an achievable goal.
Collapse
Affiliation(s)
- Laura R. Johnson
- Medical Research Council Laboratory for Molecular Cell Biology and Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - James D. Robinson
- Medical Research Council Laboratory for Molecular Cell Biology and Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Katrina N. Lester
- Medical Research Council Laboratory for Molecular Cell Biology and Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Julie A. Pitcher
- Medical Research Council Laboratory for Molecular Cell Biology and Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
43
|
Gold JI, Martini JS, Hullmann J, Gao E, Chuprun JK, Lee L, Tilley DG, Rabinowitz JE, Bossuyt J, Bers DM, Koch WJ. Nuclear translocation of cardiac G protein-Coupled Receptor kinase 5 downstream of select Gq-activating hypertrophic ligands is a calmodulin-dependent process. PLoS One 2013; 8:e57324. [PMID: 23472081 PMCID: PMC3589474 DOI: 10.1371/journal.pone.0057324] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2012] [Accepted: 01/21/2013] [Indexed: 01/08/2023] Open
Abstract
G protein-Coupled Receptors (GPCRs) kinases (GRKs) play a crucial role in regulating cardiac hypertrophy. Recent data from our lab has shown that, following ventricular pressure overload, GRK5, a primary cardiac GRK, facilitates maladaptive myocyte growth via novel nuclear localization. In the nucleus, GRK5’s newly discovered kinase activity on histone deacetylase 5 induces hypertrophic gene transcription. The mechanisms governing the nuclear targeting of GRK5 are unknown. We report here that GRK5 nuclear accumulation is dependent on Ca2+/calmodulin (CaM) binding to a specific site within the amino terminus of GRK5 and this interaction occurs after selective activation of hypertrophic Gq-coupled receptors. Stimulation of myocytes with phenylephrine or angiotensinII causes GRK5 to leave the sarcolemmal membrane and accumulate in the nucleus, while the endothelin-1 does not cause nuclear GRK5 localization. A mutation within the amino-terminus of GRK5 negating CaM binding attenuates GRK5 movement from the sarcolemma to the nucleus and, importantly, overexpression of this mutant does not facilitate cardiac hypertrophy and related gene transcription in vitro and in vivo. Our data reveal that CaM binding to GRK5 is a physiologically relevant event that is absolutely required for nuclear GRK5 localization downstream of hypertrophic stimuli, thus facilitating GRK5-dependent regulation of maladaptive hypertrophy.
Collapse
Affiliation(s)
- Jessica I. Gold
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Jeffrey S. Martini
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Jonathan Hullmann
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Erhe Gao
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - J. Kurt Chuprun
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Linda Lee
- Department of Pharmacology, University of California Davis, Davis, California, United States of America
| | - Douglas G. Tilley
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Joseph E. Rabinowitz
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Julie Bossuyt
- Department of Pharmacology, University of California Davis, Davis, California, United States of America
| | - Donald M. Bers
- Department of Pharmacology, University of California Davis, Davis, California, United States of America
| | - Walter J. Koch
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Pharmacology, Temple University School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
44
|
Suo WZ. Accelerating Alzheimer’s pathogenesis by GRK5 deficiency via cholinergic dysfunction. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/aad.2013.24020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
45
|
Kamal FA, Travers JG, Blaxall BC. G protein-coupled receptor kinases in cardiovascular disease: why "where" matters. Trends Cardiovasc Med 2012; 22:213-9. [PMID: 23062971 DOI: 10.1016/j.tcm.2012.07.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Cardiac function is mainly controlled by β-adrenergic receptors (β-ARs), members of the G protein-coupled receptor (GPCR) family. GPCR signaling and expression are tightly controlled by G protein-coupled receptor kinases (GRKs), which induce GPCR internalization and signal termination through phosphorylation. Reduced β-AR density and activity associated with elevated cardiac GRK expression and activity have been described in various cardiovascular diseases. Moreover, alterations in extracardiac GRKs have been observed in blood vessels, adrenal glands, kidneys, and fat cells. The broad tissue distribution of GPCRs and GRKs suggests that a keen appreciation of integrative physiology may drive future therapeutic development. In this review, we provide a brief summary of GRK isoforms, subcellular localization, and interacting partners that impinge directly or indirectly on the cardiovascular system. We also discuss GRK/GPCR interactions and their implications in cardiovascular pathophysiology.
Collapse
Affiliation(s)
- Fadia A Kamal
- The Heart Institute, Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | |
Collapse
|
46
|
Vandell AG, Lobmeyer MT, Gawronski BE, Langaee TY, Gong Y, Gums JG, Beitelshees AL, Turner ST, Chapman AB, Cooper-DeHoff RM, Bailey KR, Boerwinkle E, Pepine CJ, Liggett SB, Johnson JA. G protein receptor kinase 4 polymorphisms: β-blocker pharmacogenetics and treatment-related outcomes in hypertension. Hypertension 2012; 60:957-64. [PMID: 22949529 DOI: 10.1161/hypertensionaha.112.198721] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
G protein-coupled receptor kinases (GRKs) are important regulatory proteins for many G protein-coupled receptors, but little is known about GRK4 pharmacogenetics. We hypothesized that 3 nonsynonymous GRK4 single-nucleotide polymorphisms, R65L (rs2960306), A142V (rs1024323), and A486V (rs1801058), would be associated with blood pressure response to atenolol, but not hydrochlorothiazide, and would be associated with long-term cardiovascular outcomes (all-cause death, nonfatal myocardial infarction, nonfatal stroke) in participants treated with an atenolol-based versus verapamil-SR-based antihypertensive strategy. GRK4 single-nucleotide polymorphisms were genotyped in 768 hypertensive participants from the Pharmacogenomic Evaluation of Antihypertensive Responses (PEAR) trial. In whites and blacks, increasing copies of the variant 65L-142V haplotype were associated with significantly reduced atenolol-induced diastolic blood pressure lowering (-9.1±6.8 versus -6.8±7.1 versus -5.3±6.4 mm Hg in participants with 0, 1, and 2 copies of 65L-142V, respectively; P=0.0088). One thousand four hundred sixty participants with hypertension and coronary artery disease from the INternational VErapamil SR/Trandolapril STudy (INVEST) were genotyped, and variant alleles of all 3 GRK4 single-nucleotide polymorphisms were associated with increased risk for adverse cardiovascular outcomes in an additive fashion, with 486V homozygotes reaching statistical significance (odds ratio, 2.29 [1.48-3.55]; P=0.0002). These effects on adverse cardiovascular outcomes were independent of antihypertensive treatment. These results suggest that the presence of GRK4 variant alleles may be important determinants of blood pressure response to atenolol and risk for adverse cardiovascular events. The associations with GRK4 variant alleles were stronger in patients who were also ADRB1 389R homozygotes, suggesting a potential interaction between these 2 genes.
Collapse
Affiliation(s)
- Alexander G Vandell
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, University of Florida College of Pharmacy, 1600 SW Archer Rd, Room PG-22, Box 100486, Gainesville, FL 32610-0486, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Daaka Y. S-nitrosylation-regulated GPCR signaling. BIOCHIMICA ET BIOPHYSICA ACTA 2012; 1820:743-51. [PMID: 21397660 PMCID: PMC3131494 DOI: 10.1016/j.bbagen.2011.03.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Revised: 02/26/2011] [Accepted: 03/04/2011] [Indexed: 01/14/2023]
Abstract
G protein-coupled receptors (GPCRs) are the most numerous and diverse type of cell surface receptors, accounting for about 1% of the entire human genome and relaying signals from a variety of extracellular stimuli that range from lipid and peptide growth factors to ions and sensory inputs. Activated GPCRs regulate a multitude of target cell functions, including intermediary metabolism, growth and differentiation, and migration and invasion. The GPCRs contain a characteristic 7-transmembrane domain topology and their activation promotes complex formation with a variety of intracellular partner proteins, which form basis for initiation of distinct signaling networks as well as dictate fate of the receptor itself. Both termination of active GPCR signaling and removal from the plasma membrane are controlled by protein post-translational modifications of the receptor itself and its interacting partners. Phosphorylation, acylation and ubiquitination are the most studied post-translational modifications involved in GPCR signal transduction, subcellular trafficking and overall expression. Emerging evidence demonstrates that protein S-nitrosylation, the covalent attachment of a nitric oxide moiety to specified cysteine thiol groups, of GPCRs and/or their associated effectors also participates in the fine-tuning of receptor signaling and expression. This newly appreciated mode of GPCR system modification adds another set of controls to more precisely regulate the many cellular functions elicited by this large group of receptors. This article is part of a Special Issue entitled: Regulation of cellular processes by S-nitrosylation.
Collapse
Affiliation(s)
- Yehia Daaka
- The Department of Microbiology and Immunology, University of California, San Francisco, CA, United States.
| |
Collapse
|
48
|
Mushegian A, Gurevich VV, Gurevich EV. The origin and evolution of G protein-coupled receptor kinases. PLoS One 2012; 7:e33806. [PMID: 22442725 PMCID: PMC3307776 DOI: 10.1371/journal.pone.0033806] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 02/22/2012] [Indexed: 01/25/2023] Open
Abstract
G protein-coupled receptor (GPCR) kinases (GRKs) play key role in homologous desensitization of GPCRs. GRKs phosphorylate activated receptors, promoting high affinity binding of arrestins, which precludes G protein coupling. Direct binding to active GPCRs activates GRKs, so that they selectively phosphorylate only the activated form of the receptor regardless of the accessibility of the substrate peptides within it and their Ser/Thr-containing sequence. Mammalian GRKs were classified into three main lineages, but earlier GRK evolution has not been studied. Here we show that GRKs emerged at the early stages of eukaryotic evolution via an insertion of a kinase similar to ribosomal protein S6 kinase into a loop in RGS domain. GRKs in Metazoa fall into two clades, one including GRK2 and GRK3, and the other consisting of all remaining GRKs, split into GRK1-GRK7 lineage and GRK4-GRK5-GRK6 lineage in vertebrates. One representative of each of the two ancient clades is found as early as placozoan Trichoplax adhaerens. Several protists, two oomycetes and unicellular brown algae have one GRK-like protein, suggesting that the insertion of a kinase domain into the RGS domain preceded the origin of Metazoa. The two GRK families acquired distinct structural units in the N- and C-termini responsible for membrane recruitment and receptor association. Thus, GRKs apparently emerged before animals and rapidly expanded in true Metazoa, most likely due to the need for rapid signalling adjustments in fast-moving animals.
Collapse
Affiliation(s)
- Arcady Mushegian
- Stowers Institute for Medical Research, Kansas City, Missouri, United States of America
- Department of Microbiology, Kansas University Medical Center, Kansas City, Kansas, United States of America
| | - Vsevolod V. Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Eugenia V. Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, United States of America
| |
Collapse
|
49
|
Tóth DJ, Tóth JT, Gulyás G, Balla A, Balla T, Hunyady L, Várnai P. Acute depletion of plasma membrane phosphatidylinositol 4,5-bisphosphate impairs specific steps in endocytosis of the G-protein-coupled receptor. J Cell Sci 2012; 125:2185-97. [PMID: 22357943 DOI: 10.1242/jcs.097279] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Receptor endocytosis plays an important role in regulating the responsiveness of cells to specific ligands. Phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P(2)] has been shown to be crucial for endocytosis of some cell surface receptors, such as EGF and transferrin receptors, but its role in G-protein-coupled receptor internalization has not been investigated. By using luciferase-labeled type 1 angiotensin II (AT1R), type 2C serotonin (5HT2CR) or β(2) adrenergic (β2AR) receptors and fluorescently tagged proteins (β-arrestin-2, plasma-membrane-targeted Venus, Rab5) we were able to follow the sequence of molecular interactions along the endocytic route of the receptors in HEK293 cells using the highly sensitive method of bioluminescence resonance energy transfer and confocal microscopy. To study the role of plasma membrane PtdIns(4,5)P(2) in receptor endocytosis, we used our previously developed rapamycin-inducible heterodimerization system, in which the recruitment of a 5-phosphatase domain to the plasma membrane degrades PtdIns(4,5)P(2). Here we show that ligand-induced interaction of AT1, 5HT2C and β(2)A receptors with β-arrestin-2 was unaffected by PtdIns(4,5)P(2) depletion. However, trafficking of the receptors to Rab5-positive early endosomes was completely abolished in the absence of PtdIns(4,5)P(2). Remarkably, removal of the receptors from the plasma membrane was reduced but not eliminated after PtdIns(4,5)P(2) depletion. Under these conditions, stimulated AT1 receptors clustered along the plasma membrane, but did not enter the cells. Our data suggest that in the absence of PtdIns(4,5)P(2), these receptors move into clathrin-coated membrane structures, but these are not cleaved efficiently and hence cannot reach the early endosomal compartment.
Collapse
Affiliation(s)
- Dániel J Tóth
- Department of Physiology, Semmelweis University, Faculty of Medicine, Budapest, Hungary
| | | | | | | | | | | | | |
Collapse
|
50
|
Gurevich EV, Tesmer JJG, Mushegian A, Gurevich VV. G protein-coupled receptor kinases: more than just kinases and not only for GPCRs. Pharmacol Ther 2012; 133:40-69. [PMID: 21903131 PMCID: PMC3241883 DOI: 10.1016/j.pharmthera.2011.08.001] [Citation(s) in RCA: 336] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 08/01/2011] [Indexed: 12/24/2022]
Abstract
G protein-coupled receptor (GPCR) kinases (GRKs) are best known for their role in homologous desensitization of GPCRs. GRKs phosphorylate activated receptors and promote high affinity binding of arrestins, which precludes G protein coupling. GRKs have a multidomain structure, with the kinase domain inserted into a loop of a regulator of G protein signaling homology domain. Unlike many other kinases, GRKs do not need to be phosphorylated in their activation loop to achieve an activated state. Instead, they are directly activated by docking with active GPCRs. In this manner they are able to selectively phosphorylate Ser/Thr residues on only the activated form of the receptor, unlike related kinases such as protein kinase A. GRKs also phosphorylate a variety of non-GPCR substrates and regulate several signaling pathways via direct interactions with other proteins in a phosphorylation-independent manner. Multiple GRK subtypes are present in virtually every animal cell, with the highest expression levels found in neurons, with their extensive and complex signal regulation. Insufficient or excessive GRK activity was implicated in a variety of human disorders, ranging from heart failure to depression to Parkinson's disease. As key regulators of GPCR-dependent and -independent signaling pathways, GRKs are emerging drug targets and promising molecular tools for therapy. Targeted modulation of expression and/or of activity of several GRK isoforms for therapeutic purposes was recently validated in cardiac disorders and Parkinson's disease.
Collapse
Affiliation(s)
- Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University, 2200 Pierce Avenue, Preston Research Building, Rm. 454, Nashville, TN 37232, United States.
| | | | | | | |
Collapse
|