1
|
Ma P, Ou H, Cai J, Zhang Y, Ou Y. DRD2-Mediated AMPK Ubiquitination Regulates the Occurrence of Hepatic Steatosis. Liver Int 2025; 45:e70053. [PMID: 40052721 DOI: 10.1111/liv.70053] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/12/2025] [Accepted: 02/23/2025] [Indexed: 05/13/2025]
Abstract
BACKGROUND & AIMS G protein-coupled receptors (GPCRs) are important potential drug targets for the treatment of metabolic disorders. The D2 dopamine receptor (DRD2), a GPCR receptor, is a member of the dopamine receptor family. However, the role of DRD2 in regulating lipid metabolism, especially in hepatic steatosis, is unclear. METHODS Eight-week male mice were fed HFHC/MCD to induce the MASH model. AAV2/8 containing the TBG promoter was used to knock down and overexpress DRD2 in mouse liver. Co-immunoprecipitation, Western lotting, immunofluorescence, and immunohistochemistry were used to investigate the mechanisms and screen DRD2 antagonists. RESULTS The study found that activation of PKC leads to the elevation and internalisation of DRD2 in a high-fat environment. Knockdown of DRD2 in mouse liver can effectively interfere with the progression of MASH, while overexpression of DRD2 significantly aggravates the process of MASH. The study on the mechanism of DRD2 regulating lipid metabolism found that the internalisation of DRD2 could lead to dephosphorylation of pAKT (T308) by binding to β-arrestin2 and pAKT, thereby inducing ubiquitin-dependent degradation of AMPK and exacerbating steatosis. L-741626, a DRD2 antagonist, was found to interfere with the internalisation of DRD2 in a high-fat environment. It has been shown that L-741626 can treat MASH by regulating the AKT-AMPK signalling axis in vitro and in vivo. CONCLUSIONS In conclusion, this study demonstrated that internalisation of DRD2 in a high-fat environment aggravated MASH progression through the AKT-AMPK signalling axis. Furthermore, L-741626, as a DRD2 antagonist, has the potential to treat MASH.
Collapse
Affiliation(s)
- Peng Ma
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Hao Ou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Junze Cai
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yuanli Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yu Ou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Ouaidat S, Amaral IM, Monteiro DG, Harati H, Hofer A, El Rawas R. Orexins/Hypocretins: Gatekeepers of Social Interaction and Motivation. Int J Mol Sci 2024; 25:2609. [PMID: 38473854 PMCID: PMC10931973 DOI: 10.3390/ijms25052609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/12/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Ever since the discovery of the brain's orexin/hypocretin system, most research was directed toward unveiling its contribution to the normal functioning of individuals. The investigation of reward-seeking behaviors then gained a lot of attention once the distribution of orexinergic neurons was revealed. Here, we discuss findings on the involvement of orexins in social interaction, a natural reward type. While some studies have succeeded in defining the relationship between orexin and social interaction, the controversy regarding its nature (direct or inverse relation) raises questions about what aspects have been overlooked until now. Upon examining the literature, we identified a research gap concerning conditions influencing the impact of orexins on social behavior expression. In this review, we introduce a number of factors (e.g., stress, orexin's source) that must be considered while studying the role of orexins in social interaction. Furthermore, we refer to published research to investigate the stage at which orexins affect social interaction and we highlight the nucleus accumbens (NAc) shell's role in social interaction and other rewarding behaviors. Finally, the underlying orexin molecular pathway influencing social motivation in particular illnesses is proposed. We conclude that orexin's impact on social interaction is multifactorial and depends on specific conditions available at a time.
Collapse
Affiliation(s)
- Sara Ouaidat
- Division of Psychiatry I, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, 6020 Innsbruck, Austria
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut P.O. Box 1533, Lebanon
| | - Inês M. Amaral
- Division of Psychiatry I, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Diogo G. Monteiro
- Division of Psychiatry I, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Hayat Harati
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut P.O. Box 1533, Lebanon
| | - Alex Hofer
- Division of Psychiatry I, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Rana El Rawas
- Division of Psychiatry I, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
3
|
Sánchez-Soto M, Boldizsar NM, Schardien KA, Madaras NS, Willette BKA, Inbody LR, Dasaro C, Moritz AE, Drube J, Haider RS, Free RB, Hoffman C, Sibley DR. G Protein-Coupled Receptor Kinase 2 Selectively Enhances β-Arrestin Recruitment to the D 2 Dopamine Receptor through Mechanisms That Are Independent of Receptor Phosphorylation. Biomolecules 2023; 13:1552. [PMID: 37892234 PMCID: PMC10605370 DOI: 10.3390/biom13101552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
The D2 dopamine receptor (D2R) signals through both G proteins and β-arrestins to regulate important physiological processes, such as movement, reward circuitry, emotion, and cognition. β-arrestins are believed to interact with G protein-coupled receptors (GPCRs) at the phosphorylated C-terminal tail or intracellular loops. GPCR kinases (GRKs) are the primary drivers of GPCR phosphorylation, and for many receptors, receptor phosphorylation is indispensable for β-arrestin recruitment. However, GRK-mediated receptor phosphorylation is not required for β-arrestin recruitment to the D2R, and the role of GRKs in D2R-β-arrestin interactions remains largely unexplored. In this study, we used GRK knockout cells engineered using CRISPR-Cas9 technology to determine the extent to which β-arrestin recruitment to the D2R is GRK-dependent. Genetic elimination of all GRK expression decreased, but did not eliminate, agonist-stimulated β-arrestin recruitment to the D2R or its subsequent internalization. However, these processes were rescued upon the re-introduction of various GRK isoforms in the cells with GRK2/3 also enhancing dopamine potency. Further, treatment with compound 101, a pharmacological inhibitor of GRK2/3 isoforms, decreased β-arrestin recruitment and receptor internalization, highlighting the importance of this GRK subfamily for D2R-β-arrestin interactions. These results were recapitulated using a phosphorylation-deficient D2R mutant, emphasizing that GRKs can enhance β-arrestin recruitment and activation independently of receptor phosphorylation.
Collapse
Affiliation(s)
- Marta Sánchez-Soto
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, Bethesda, MD 20892, USA (R.B.F.)
| | - Noelia M. Boldizsar
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, Bethesda, MD 20892, USA (R.B.F.)
| | - Kayla A. Schardien
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, Bethesda, MD 20892, USA (R.B.F.)
| | - Nora S. Madaras
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, Bethesda, MD 20892, USA (R.B.F.)
| | - Blair K. A. Willette
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, Bethesda, MD 20892, USA (R.B.F.)
| | - Laura R. Inbody
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, Bethesda, MD 20892, USA (R.B.F.)
| | - Christopher Dasaro
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, Bethesda, MD 20892, USA (R.B.F.)
| | - Amy E. Moritz
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, Bethesda, MD 20892, USA (R.B.F.)
| | - Julia Drube
- Institut für Molekulare Zellbiologie, CMB-Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745 Jena, Germany (R.S.H.); (C.H.)
| | - Raphael S. Haider
- Institut für Molekulare Zellbiologie, CMB-Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745 Jena, Germany (R.S.H.); (C.H.)
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK
- Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, Birmingham B15 2TT, UK
| | - R. Benjamin Free
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, Bethesda, MD 20892, USA (R.B.F.)
| | - Carsten Hoffman
- Institut für Molekulare Zellbiologie, CMB-Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, D-07745 Jena, Germany (R.S.H.); (C.H.)
| | - David R. Sibley
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, Bethesda, MD 20892, USA (R.B.F.)
| |
Collapse
|
4
|
Bertran-Gonzalez J, Dinale C, Matamales M. Restoring the youthful state of striatal plasticity in aged mice re-enables cognitive control of action. Curr Biol 2023; 33:1997-2007.e5. [PMID: 37141886 DOI: 10.1016/j.cub.2023.04.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/21/2023] [Accepted: 04/12/2023] [Indexed: 05/06/2023]
Abstract
Multidisciplinary evidence suggests that the control of voluntary action arbitrates between two major forms of behavioral processing: cognitively guided (or goal directed) and autonomously guided (or habitual). Brain-state irregularities affecting the striatum-such as aging-commonly shift control toward the latter, although the responsible neural mechanisms remain unknown. Combining instrumental conditioning with cell-specific mapping and chemogenetics in striatal neurons, we explored strategies that invigorate goal-directed capacity in aged mice. We found that, under conditions favoring goal-directed control, aged animals resiliently expressed autonomously guided behavior, a response that was underpinned by a characteristic one-to-one functional engagement of the two main neuronal populations in the striatum-D1- and D2-dopamine receptor-expressing spiny projection neurons (SPNs). Chemogenetically induced desensitization of D2-SPN signaling in aged transgenic mice recapitulated the striatal plasticity state observed in young mice, an effect that shifted behavior toward vigorous, goal-directed action. Our findings contribute to the understanding of the neural bases of behavioral control and propose neural system interventions that enhance cognitive functioning in habit-prone brains.
Collapse
Affiliation(s)
- Jesus Bertran-Gonzalez
- Decision Neuroscience Laboratory, School of Psychology, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Caroline Dinale
- Decision Neuroscience Laboratory, School of Psychology, University of New South Wales, Sydney, NSW 2052, Australia
| | - Miriam Matamales
- Decision Neuroscience Laboratory, School of Psychology, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
5
|
Kim KM. Unveiling the Differences in Signaling and Regulatory Mechanisms between Dopamine D2 and D3 Receptors and Their Impact on Behavioral Sensitization. Int J Mol Sci 2023; 24:ijms24076742. [PMID: 37047716 PMCID: PMC10095578 DOI: 10.3390/ijms24076742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 04/09/2023] Open
Abstract
Dopamine receptors are classified into five subtypes, with D2R and D3R playing a crucial role in regulating mood, motivation, reward, and movement. Whereas D2R are distributed widely across the brain, including regions responsible for motor functions, D3R are primarily found in specific areas related to cognitive and emotional functions, such as the nucleus accumbens, limbic system, and prefrontal cortex. Despite their high sequence homology and similar signaling pathways, D2R and D3R have distinct regulatory properties involving desensitization, endocytosis, posttranslational modification, and interactions with other cellular components. In vivo, D3R is closely associated with behavioral sensitization, which leads to increased dopaminergic responses. Behavioral sensitization is believed to result from D3R desensitization, which removes the inhibitory effect of D3R on related behaviors. Whereas D2R maintains continuous signal transduction through agonist-induced receptor phosphorylation, arrestin recruitment, and endocytosis, which recycle and resensitize desensitized receptors, D3R rarely undergoes agonist-induced endocytosis and instead is desensitized after repeated agonist exposure. In addition, D3R undergoes more extensive posttranslational modifications, such as glycosylation and palmitoylation, which are needed for its desensitization. Overall, a series of biochemical settings more closely related to D3R could be linked to D3R-mediated behavioral sensitization.
Collapse
Affiliation(s)
- Kyeong-Man Kim
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 61186, Republic of Korea
| |
Collapse
|
6
|
Xu J, Pittenger C. The histamine H3 receptor modulates dopamine D2 receptor-dependent signaling pathways and mouse behaviors. J Biol Chem 2023; 299:104583. [PMID: 36871761 PMCID: PMC10139999 DOI: 10.1016/j.jbc.2023.104583] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
The histamine H3 receptor (H3R) is highly enriched in the spiny projection neurons (SPNs) of the striatum, in both the D1 receptor (D1R)-expressing and D2 receptor (D2R)-expressing populations. A crossantagonistic interaction between H3R and D1R has been demonstrated in mice, both at the behavioral level and at the biochemical level. Although interactive behavioral effects have been described upon coactivation of H3R and D2R, the molecular mechanisms underlying this interaction are poorly understood. Here, we show that activation of H3R with the selective agonist R-(-)-α-methylhistamine dihydrobromide mitigates D2R agonist-induced locomotor activity and stereotypic behavior. Using biochemical approaches and the proximity ligation assay, we demonstrated the existence of an H3R-D2R complex in the mouse striatum. In addition, we examined consequences of simultaneous H3R-D2R agonism on the phosphorylation levels of several signaling molecules using immunohistochemistry. H3R agonist treatment modulated Akt (serine/threonine PKB)-glycogen synthase kinase 3 beta signaling in response to D2R activation via a β-arrestin 2-dependent mechanism in D2R-SPNs but not in D1R-SPNs. Phosphorylation of mitogen- and stress-activated protein kinase 1 and rpS6 (ribosomal protein S6) was largely unchanged under these conditions. As Akt-glycogen synthase kinase 3 beta signaling has been implicated in several neuropsychiatric disorders, this work may help clarify the role of H3R in modulating D2R function, leading to a better understanding of pathophysiology involving the interaction between histamine and dopamine systems.
Collapse
Affiliation(s)
- Jian Xu
- Department of Psychiatry, Yale University. ,
| | - Christopher Pittenger
- Department of Psychiatry, Yale University; Department of Psychology, Yale University; Department of Child Study Center, Yale University; Department of Interdepartmental Neuroscience Program, Yale University; Department of Wu-Tsai Institute, Yale University; Department of Center for Brain and Mind Health, Yale University.
| |
Collapse
|
7
|
Ubeysinghe S, Wijayaratna D, Kankanamge D, Karunarathne A. Molecular regulation of PLCβ signaling. Methods Enzymol 2023; 682:17-52. [PMID: 36948701 PMCID: PMC11863860 DOI: 10.1016/bs.mie.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Phospholipase C (PLC) enzymes convert the membrane phospholipid phosphatidylinositol-4,5-bisphosphate (PIP2) into inositol-1,4,5-triphosphate (IP3) and diacylglycerol (DAG). IP3 and DAG regulate numerous downstream pathways, eliciting diverse and profound cellular changes and physiological responses. In the six PLC subfamilies in higher eukaryotes, PLCβ is intensively studied due to its prominent role in regulating crucial cellular events underlying many processes including cardiovascular and neuronal signaling, and associated pathological conditions. In addition to GαqGTP, Gβγ generated upon G protein heterotrimer dissociation also regulates PLCβ activity. Here, we not only review how Gβγ directly activates PLCβ, and also extensively modulates Gαq-mediated PLCβ activity, but also provide a structure-function overview of PLC family members. Given that Gαq and PLCβ are oncogenes, and Gβγ shows unique cell-tissue-organ specific expression profiles, Gγ subtype-dependent signaling efficacies, and distinct subcellular activities, this review proposes that Gβγ is a major regulator of Gαq-dependent and independent PLCβ signaling.
Collapse
Affiliation(s)
| | | | - Dinesh Kankanamge
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Ajith Karunarathne
- Department of Chemistry, St. Louis University, St. Louis, MO, United States.
| |
Collapse
|
8
|
Kearney PJ, Bolden NC, Kahuno E, Conklin TL, Martin GE, Lubec G, Melikian HE. Presynaptic Gq-coupled receptors drive biphasic dopamine transporter trafficking that modulates dopamine clearance and motor function. J Biol Chem 2023; 299:102900. [PMID: 36640864 PMCID: PMC9943899 DOI: 10.1016/j.jbc.2023.102900] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/23/2022] [Accepted: 01/03/2023] [Indexed: 01/13/2023] Open
Abstract
Extracellular dopamine (DA) levels are constrained by the presynaptic DA transporter (DAT), a major psychostimulant target. Despite its necessity for DA neurotransmission, DAT regulation in situ is poorly understood, and it is unknown whether regulated DAT trafficking impacts dopaminergic signaling and/or behaviors. Leveraging chemogenetics and conditional gene silencing, we found that activating presynaptic Gq-coupled receptors, either hM3Dq or mGlu5, drove rapid biphasic DAT membrane trafficking in ex vivo striatal slices, with region-specific differences between ventral and dorsal striata. DAT insertion required D2 DA autoreceptors and intact retromer, whereas DAT retrieval required PKC activation and Rit2. Ex vivo voltammetric studies revealed that DAT trafficking impacts DA clearance. Furthermore, dopaminergic mGlu5 silencing elevated DAT surface expression and abolished motor learning, which was rescued by inhibiting DAT with a subthreshold CE-158 dose. We discovered that presynaptic DAT trafficking is complex, multimodal, and region specific, and for the first time, we identified cell autonomous mechanisms that govern presynaptic DAT tone. Importantly, the findings are consistent with a role for regulated DAT trafficking in DA clearance and motor function.
Collapse
Affiliation(s)
- Patrick J. Kearney
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA,Morningside Graduate School of Biomedical Sciences, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Nicholas C. Bolden
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA,Morningside Graduate School of Biomedical Sciences, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Elizabeth Kahuno
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Tucker L. Conklin
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Gilles E. Martin
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA
| | - Gert Lubec
- Department of Neuroproteomics, Paracelsus Private Medical University, Salzburg, Austria
| | - Haley E. Melikian
- Brudnick Neuropsychiatric Research Institute, Department of Neurobiology, UMASS Chan Medical School, Worcester, Massachusetts, USA,For correspondence: Haley E. Melikian
| |
Collapse
|
9
|
Ågren R, Sahlholm K. G protein-coupled receptor kinase-2 confers isoform-specific calcium sensitivity to dopamine D 2 receptor desensitization. FASEB J 2021; 35:e22013. [PMID: 34699610 DOI: 10.1096/fj.202100704rr] [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: 04/26/2021] [Revised: 10/04/2021] [Accepted: 10/11/2021] [Indexed: 11/11/2022]
Abstract
The dopamine D2 receptor (D2 R) functions as an autoreceptor on dopaminergic cell bodies and terminals and as a postsynaptic receptor on a variety of neurons in the central nervous system. As a result of alternative splicing, the D2 R is expressed as two isoforms: long (D2L R) and short (D2S R) differing by a stretch of 29 residues in the third intracellular loop, with D2S R being the predominant presynaptic isoform. Recent reports described a Ca2+ sensitivity of the desensitization time course of potassium currents elicited via D2S R, but not via D2L R, when either isoform was selectively expressed in dopaminergic neurons. Here, we aimed to study the mechanism behind this subtype-specific Ca2+ sensitivity. Thus, we measured the desensitization of potassium channel responses evoked by D2L R and D2S R using two-electrode voltage clamp in Xenopus oocytes in the absence and presence of different amounts of β-arrestin2 and G protein-coupled receptor kinase-2 (GRK2), both of which are known to play important roles in D2 R desensitization in native cells. We found that co-expression of both GRK2 and β-arrestin2 was necessary for reconstitution of the Ca2+ sensitivity of D2S R desensitization, while D2L R did not display Ca2+ sensitivity under these conditions. The effect of Ca2+ chelation by BAPTA-AM to slow the rate of D2S R desensitization was mimicked by the GRK2 inhibitor, Cmpd101, and by the kinase-inactivating GRK2 mutation, K220R, but not by the PKC inhibitor, Gö6976, nor by the calmodulin antagonist, KN-93. Thus, Ca2+ -sensitive desensitization of D2S R appears to be mediated via a GRK2 phosphorylation-dependent mechanism.
Collapse
Affiliation(s)
- Richard Ågren
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Kristoffer Sahlholm
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Integrative Medical Biology, Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
10
|
Mann A, Keen AC, Mark H, Dasgupta P, Javitch JA, Canals M, Schulz S, Robert Lane J. New phosphosite-specific antibodies to unravel the role of GRK phosphorylation in dopamine D 2 receptor regulation and signaling. Sci Rep 2021; 11:8288. [PMID: 33859231 PMCID: PMC8050214 DOI: 10.1038/s41598-021-87417-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/25/2021] [Indexed: 12/20/2022] Open
Abstract
The dopamine D2 receptor (D2R) is the target of drugs used to treat the symptoms of Parkinson’s disease and schizophrenia. The D2R is regulated through its interaction with and phosphorylation by G protein receptor kinases (GRKs) and interaction with arrestins. More recently, D2R arrestin-mediated signaling has been shown to have distinct physiological functions to those of G protein signalling. Relatively little is known regarding the patterns of D2R phosphorylation that might control these processes. We aimed to generate antibodies specific for intracellular D2R phosphorylation sites to facilitate the investigation of these mechanisms. We synthesised double phosphorylated peptides corresponding to regions within intracellular loop 3 of the hD2R and used them to raise phosphosite-specific antibodies to capture a broad screen of GRK-mediated phosphorylation. We identify an antibody specific to a GRK2/3 phosphorylation site in intracellular loop 3 of the D2R. We compared measurements of D2R phosphorylation with other measurements of D2R signalling to profile selected D2R agonists including previously described biased agonists. These studies demonstrate the utility of novel phosphosite-specific antibodies to investigate D2R regulation and signalling.
Collapse
Affiliation(s)
- Anika Mann
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Alastair C Keen
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.,Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK.,Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK
| | - Hanka Mark
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Pooja Dasgupta
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Jonathan A Javitch
- Departments of Psychiatry and Pharmacology, Vagelos College of Physicians and Surgeons, Columbia University, New York, USA.,Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, USA
| | - Meritxell Canals
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK.,Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK
| | - Stefan Schulz
- Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany.
| | - J Robert Lane
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK. .,Centre of Membrane Proteins and Receptors, University of Birmingham and University of Nottingham, Midlands, UK.
| |
Collapse
|
11
|
Pivonello C, Patalano R, Negri M, Pirchio R, Colao A, Pivonello R, Auriemma RS. Resistance to Dopamine Agonists in Pituitary Tumors: Molecular Mechanisms. Front Endocrinol (Lausanne) 2021; 12:791633. [PMID: 35095761 PMCID: PMC8789681 DOI: 10.3389/fendo.2021.791633] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/16/2021] [Indexed: 11/24/2022] Open
Abstract
Pituitary neuroendocrine tumors (PitNET) are commonly benign tumors accounting for 10-25% of intracranial tumors. Prolactin-secreting adenomas represent the most predominant type of all PitNET and for this subtype of tumors, the medical therapy relies on the use of dopamine agonists (DAs). DAs yield an excellent therapeutic response in reducing tumor size and hormonal secretion targeting the dopamine receptor type 2 (D2DR) whose higher expression in prolactin-secreting adenomas compared to other PitNET is now well established. Moreover, although DAs therapy does not represent the first-line therapy for other PitNET, off-label use of DAs is considered in PitNET expressing D2DR. Nevertheless, DAs primary or secondary resistance, occurring in a subset of patients, may involve several molecular mechanisms, presently not fully elucidated. Dopamine receptors (DRs) expression is a prerequisite for a proper DA function in PitNET and several molecular events may negatively modify DR membrane expression, through the DRs down-regulation and intracellular trafficking, and DR signal transduction pathway. The current mini-review will summarise the presently known molecular events that underpin the unsuccessful therapy with DAs.
Collapse
Affiliation(s)
- Claudia Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università di Napoli (Federico II), Naples, Italy
- *Correspondence: Claudia Pivonello, ;
| | - Roberta Patalano
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università di Napoli (Federico II), Naples, Italy
- Dipartimento di Sanità Pubblica, Università di Napoli (Federico II), Naples, Italy
| | - Mariarosaria Negri
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università di Napoli (Federico II), Naples, Italy
| | - Rosa Pirchio
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università di Napoli (Federico II), Naples, Italy
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università di Napoli (Federico II), Naples, Italy
- United Nations Educational, Scientific and Cultural Organization (UNESCO) Chair for Health Education and Sustainable Development, Federico II University, Naples, Italy
| | - Rosario Pivonello
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università di Napoli (Federico II), Naples, Italy
- United Nations Educational, Scientific and Cultural Organization (UNESCO) Chair for Health Education and Sustainable Development, Federico II University, Naples, Italy
| | - Renata Simona Auriemma
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università di Napoli (Federico II), Naples, Italy
| |
Collapse
|
12
|
Novick AM, Scott AT, Neill Epperson C, Schneck CD. Neuropsychiatric effects of tamoxifen: Challenges and opportunities. Front Neuroendocrinol 2020; 59:100869. [PMID: 32822707 PMCID: PMC7669724 DOI: 10.1016/j.yfrne.2020.100869] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 08/12/2020] [Accepted: 08/16/2020] [Indexed: 02/08/2023]
Abstract
Epidemiological, clinical, and basic research over the past thirty years have described the benefits of estrogen on cognition, mood, and brain health. Less is known about tamoxifen, a selective estrogen receptor modifier (SERM) commonly used in breast cancer which is able to cross the blood-brain barrier. In this article, we review the basic pharmacology of tamoxifenas well as its effects on cognition and mood. The literature reveals an overall impairing effect of tamoxifen on cognition in breast cancer patients, hinting at central antiestrogen activity. On the other hand, tamoxifen demonstrates promising effects in psychiatric disorders, like bipolar disorder, where its therapeutic action may be independent of interaction with estrogen receptors. Understanding the neuropsychiatric properties of SERMs like tamoxifen can guide future research to ameliorate unwanted side-effects and provide novel options for difficult to treat disorders.
Collapse
Affiliation(s)
- Andrew M Novick
- Department of Psychiatry, University of Colorado School of Medicine, 13001 E 17th Place, Campus Box F546, Aurora, CO 80045, United States.
| | - Anthony T Scott
- Department of Psychiatry, University of Colorado School of Medicine, 13001 E 17th Place, Campus Box F546, Aurora, CO 80045, United States
| | - C Neill Epperson
- Department of Psychiatry, University of Colorado School of Medicine, 13001 E 17th Place, Campus Box F546, Aurora, CO 80045, United States
| | - Christopher D Schneck
- Department of Psychiatry, University of Colorado School of Medicine, 13001 E 17th Place, Campus Box F546, Aurora, CO 80045, United States
| |
Collapse
|
13
|
Chen R, Ferris MJ, Wang S. Dopamine D2 autoreceptor interactome: Targeting the receptor complex as a strategy for treatment of substance use disorder. Pharmacol Ther 2020; 213:107583. [PMID: 32473160 PMCID: PMC7434700 DOI: 10.1016/j.pharmthera.2020.107583] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 05/11/2020] [Indexed: 02/06/2023]
Abstract
Dopamine D2 autoreceptors (D2ARs), located in somatodendritic and axon terminal compartments of dopamine (DA) neurons, function to provide a negative feedback regulatory control on DA neuron firing, DA synthesis, reuptake and release. Dysregulation of D2AR-mediated DA signaling is implicated in vulnerability to substance use disorder (SUD). Due to the extreme low abundance of D2ARs compared to postsynaptic D2 receptors (D2PRs) and the lack of experimental tools to differentiate the signaling of D2ARs from D2PRs, the regulation of D2ARs by drugs of abuse is poorly understood. The recent availability of conditional D2AR knockout mice and newly developed virus-mediated gene delivery approaches have provided means to specifically study the function of D2ARs at the molecular, cellular and behavioral levels. There is a growing revelation of novel mechanisms and new proteins that mediate D2AR activity, suggesting that D2ARs act cooperatively with an array of membrane and intracellular proteins to tightly control DA transmission. This review highlights D2AR-interacting partners including transporters, G-protein-coupled receptors, ion channels, intracellular signaling modulators, and protein kinases. The complexity of the D2AR interaction network illustrates the functional divergence of D2ARs. Pharmacological targeting of multiple D2AR-interacting partners may be more effective to restore disrupted DA homeostasis by drugs of abuse.
Collapse
Affiliation(s)
- Rong Chen
- Dept. of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America; Center for the Neurobiology of Addiction Treatment, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America.
| | - Mark J Ferris
- Dept. of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America; Center for the Neurobiology of Addiction Treatment, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America
| | - Shiyu Wang
- Dept. of Physiology & Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, United States of America
| |
Collapse
|
14
|
He X, Yan L, Wu Q, Zhang G, Zhou N. Ligand-dependent internalization of Bombyx mori tachykinin-related peptide receptor is regulated by PKC, GRK5 and β-arrestin2/BmKurtz. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118690. [PMID: 32112783 DOI: 10.1016/j.bbamcr.2020.118690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 02/18/2020] [Accepted: 02/23/2020] [Indexed: 10/24/2022]
Abstract
Tachykinin signaling system is present in both vertebrates and invertebrates, and functions as neuromodulator responsible for the regulation of various physiological processes. In human, the internalization of G protein-coupled receptors has been extensively characterized; however, the insect GPCR internalization has been rarely investigated. Here, we constructed two expression vectors of Bombyx tachykinin-related peptide receptor (BmTKRPR) fused with Enhanced Green Fluorescent Protein (EGFP) at the C-terminal end for direct visualization of receptor expression, localization, and trafficking in cultured mammalian HEK293 and insect Sf21 cells. Our results demonstrated that agonist-activated BmTKRPR underwent rapid internalization in a dose-and time-dependent manner via a clathrin-dependent pathway in both HEK293 and Sf21 cells. Further investigation via RNAi or specific inhibitors, or co-immunoprecipitation demonstrated that agonist-induced BmTKRPR internalization was mediated by PKC, GRK5 and β-arrestin2/BmKurtz. In addition, we also observed that most of the internalized BmTKRP receptors were recycled to the cell surface via early endosomes upon peptide ligand removal. Our study provides the first in-depth information on mechanisms underlying insect TKRP receptor internalization and perhaps aids in the interpretation of the signaling in the regulation of physiological processes.
Collapse
Affiliation(s)
- Xiaobai He
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China; College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212018, China.
| | - Lili Yan
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Qi Wu
- College of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212018, China
| | - Guozheng Zhang
- Key Laboratory of Genetic Improvement of Sericulture, Ministry of Agriculture and Rural Affairs, Jiangsu University of Science and Technology, Zhenjiang, Jiangsu 212018, China
| | - Naiming Zhou
- Institute of Biochemistry, College of Life Sciences, Zijingang Campus, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
15
|
He D, Lasek AW. Anaplastic Lymphoma Kinase Regulates Internalization of the Dopamine D2 Receptor. Mol Pharmacol 2020; 97:123-131. [PMID: 31734646 PMCID: PMC6964149 DOI: 10.1124/mol.119.117473] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 11/11/2019] [Indexed: 12/16/2022] Open
Abstract
The dopamine D2 receptor (D2R) is a G protein-coupled receptor (GPCR) expressed in regions of the brain that control motor function, cognition, and motivation. As a result, D2R is involved in the pathophysiology of disorders such as schizophrenia and drug addiction. Understanding the signaling pathways activated by D2R is crucial to finding new therapeutic targets for these disorders. D2R stimulation by its agonist, dopamine, causes desensitization and internalization of the receptor. A previous study found that inhibitors of the receptor tyrosine kinase anaplastic lymphoma kinase (ALK) blocked D2R desensitization in neurons in the ventral tegmental area of the brain. In the present study, using a cell-based system, we investigated whether ALK regulates D2R internalization. The ALK inhibitor alectinib completely inhibited dopamine-induced D2R internalization. Since GPCRs can transactivate receptor tyrosine kinases, we also examined if D2R stimulation activated ALK signaling. ALK phosphorylation increased by almost 2-fold after dopamine treatment and ALK coimmunoprecipitated with D2R. To identify the signaling pathways downstream of ALK that might regulate D2R internalization, we used pharmacological inhibitors of proteins activated by ALK signaling. Protein kinase Cγ was activated by dopamine in an ALK-dependent manner, and a protein kinase C inhibitor completely blocked dopamine-induced D2R internalization. Taken together, these results identify ALK as a receptor tyrosine kinase transactivated by D2R that promotes its internalization, possibly through activation of protein kinase C. ALK inhibitors could be useful in enhancing D2R signaling. SIGNIFICANCE STATEMENT: Receptor internalization is a mechanism by which receptors are desensitized. In this study we found that agonist-induced internalization of the dopamine D2 receptor is regulated by the receptor tyrosine kinase ALK. ALK was also transactivated by and associated with dopamine D2 receptor. Dopamine activated protein kinase C in an ALK-dependent manner and a PKC inhibitor blocked dopamine D2 receptor internalization. These results indicate that ALK regulates dopamine D2 receptor trafficking, which has implications for psychiatric disorders involving dysregulated dopamine signaling.
Collapse
Affiliation(s)
- Donghong He
- Center for Alcohol Research in Epigenetics and Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois
| | - Amy W Lasek
- Center for Alcohol Research in Epigenetics and Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
16
|
Su M, Zhou J, Duan Z, Zhang J. Transcriptional analysis of renal dopamine-mediated Na + homeostasis response to environmental salinity stress in Scatophagus argus. BMC Genomics 2019; 20:418. [PMID: 31126236 PMCID: PMC6534869 DOI: 10.1186/s12864-019-5795-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 05/10/2019] [Indexed: 02/07/2023] Open
Abstract
Background To control the osmotic pressure in the body, physiological adjustments to salinity fluctuations require the fish to regulate body fluid homeostasis in relation to environmental change via osmoregulation. Previous studies related to osmoregulation were focused primarily on the gill; however, little is known about another organ involved in osmoregulation, the kidney. The salinity adaptation of marine fish involves complex physiological traits, metabolic pathways and molecular and gene networks in osmoregulatory organs. To further explore of the salinity adaptation of marine fish with regard to the role of the kidney, the euryhaline fish Scatophagus argus was employed in the present study. Renal expression profiles of S. argus at different salinity levels were characterized using RNA-sequencing, and an integrated approach of combining molecular tools with physiological and biochemical techniques was utilized to reveal renal osmoregulatory mechanisms in vivo and in vitro. Results S. argus renal transcriptomes from the hyposaline stress (0‰, freshwater [FW]), hypersaline stress (50‰, hypersaline water [HW]) and control groups (25‰) were compared to elucidate potential osmoregulatory mechanisms. In total, 19,012 and 36,253 differentially expressed genes (DEGs) were obtained from the FW and HW groups, respectively. Based on the functional classification of DEGs, the renal dopamine system-induced Na+ transport was demonstrated to play a fundamental role in osmoregulation. In addition, for the first time in fish, many candidate genes associated with the dopamine system were identified. Furthermore, changes in environmental salinity affected renal dopamine release/reuptake by regulating the expression of genes related to dopamine reuptake (dat and nkaα1), vesicular traffic-mediated dopamine release (pink1, lrrk2, ace and apn), DAT phosphorylation (CaMKIIα and pkcβ) and internalization (akt1). The associated transcriptional regulation ensured appropriate extracellular dopamine abundance in the S. argus kidney, and fluctuations in extracellular dopamine produced a direct influence on Na+/K+-ATPase (NKA) expression and activity, which is associated with Na+ homeostasis. Conclusions These transcriptomic data provided insight into the molecular basis of renal osmoregulation in S. argus. Significantly, the results of this study revealed the mechanism of renal dopamine system-induced Na+ transport is essential in fish osmoregulation. Electronic supplementary material The online version of this article (10.1186/s12864-019-5795-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maoliang Su
- Shenzhen Key Laboratory of Marine Bioresource & Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China.,Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Jianan Zhou
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Sciences, Shanghai Ocean University, Shanghai, 201306, China
| | - Zhengyu Duan
- Shenzhen Key Laboratory of Marine Bioresource & Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China.,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Sciences, Shanghai Ocean University, Shanghai, 201306, China
| | - Junbin Zhang
- Shenzhen Key Laboratory of Marine Bioresource & Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China. .,Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
17
|
Murakami T, Enjoji M, Koyama S. Leptin attenuates D 2 receptor-mediated inhibition of putative ventral tegmental area dopaminergic neurons. Physiol Rep 2019; 6:e13631. [PMID: 29611323 PMCID: PMC5880875 DOI: 10.14814/phy2.13631] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 01/30/2018] [Accepted: 02/02/2018] [Indexed: 01/18/2023] Open
Abstract
Obesity causes hyperleptinemia. We have previously shown that D2 receptor‐mediated inhibition of ventral tegmental area (VTA) dopaminergic neurons is attenuated in diet‐induced mice with obesity. Consequently, we hypothesized that high concentrations of serum leptin during obesity might modulate D2 receptor‐mediated effects on VTA dopaminergic neurons. To investigate our hypothesis, we examined leptin effects on D2 receptor‐mediated inhibition of putative VTA dopaminergic neurons from lean mice using electrophysiological techniques. Leptin (100 nmol/L) directly inhibited spontaneous firing in 71% of putative VTA dopaminergic neurons (leptin‐responsive), whereas the remaining 29% of neurons were leptin‐nonresponsive. In 41% of leptin‐responsive neurons, leptin attenuated the reduced firing rate produced by quinpirole (100 nmol/L), whereas the remaining 59% of neurons exhibited no effect of leptin. In leptin‐nonresponsive neurons, no significant leptin‐induced effect was observed on reduced firing rate produced by quinpirole. In leptin‐responsive neurons with positive leptin‐induced attenuation of quinpirole effects, leptin‐induced attenuation persisted for >20 min, whereas no such persistent attenuation was observed in other types of neurons. In conclusion, leptin attenuates D2 receptor‐mediated inhibition in a subpopulation of putative VTA dopaminergic neurons. We suggest that leptin directly decreases, and indirectly increases, excitability of VTA dopaminergic neurons. In turn, this may contribute to a change in feeding behavior through the mesolimbic dopaminergic system during the development of obesity.
Collapse
Affiliation(s)
- Takami Murakami
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Munechika Enjoji
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | - Susumu Koyama
- Department of Clinical Pharmacology, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan.,Department of Advanced Pharmacology, Daiichi University of Pharmacy, Fukuoka, Japan
| |
Collapse
|
18
|
Zestos AG, Carpenter C, Kim Y, Low MJ, Kennedy RT, Gnegy ME. Ruboxistaurin Reduces Cocaine-Stimulated Increases in Extracellular Dopamine by Modifying Dopamine-Autoreceptor Activity. ACS Chem Neurosci 2019; 10:1960-1969. [PMID: 30384585 DOI: 10.1021/acschemneuro.8b00259] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Cocaine is a highly abused drug, and cocaine addiction affects millions of individuals worldwide. Cocaine blocks normal uptake function at the dopamine transporter (DAT), thus increasing extracellular dopamine. Currently, no chemical therapies are available to treat cocaine abuse. Previous works showed that the selective inhibitors of protein kinase Cβ (PKCβ), enzastaurin and ruboxistaurin, attenuate dopamine overflow and locomotion stimulated by another psychostimulant drug, amphetamine. We now test if ruboxistaurin similarly affects cocaine action. Perfusion of 1 μM ruboxistaurin directly into the core of the nucleus accumbens via retrodialysis reduced cocaine-stimulated increases in dopamine overflow, measured using microdialysis sampling, with simultaneous reductions in locomotor behavior. Because cocaine activity is highly regulated by dopamine autoreceptors, we examined whether ruboxistaurin was acting at the level of the D2 autoreceptor. Perfusion of 5 μM raclopride, a selective D2-like receptor antagonist, before addition of ruboxistaurin, abrogated the effect of ruboxistaurin on cocaine-stimulated dopamine overflow and hyperlocomotion. Further, ruboxistaurin was inactive against cocaine-stimulated locomotor activity in mice with a genetic deletion in D2 receptors as compared to wild-type mice. In contrast, blockade or deletion of dopamine D2 receptors did not abolish the attenuating effect of ruboxistaurin on amphetamine-stimulated activities. Therefore, the inhibition of PKCβ reduces dopamine overflow and locomotor activity stimulated by both cocaine and amphetamine, but the mechanism of action differs for each stimulant. These data suggest that inhibition of PKCβ would serve as a target to reduce the abuse of either amphetamine or cocaine.
Collapse
Affiliation(s)
- Alexander G. Zestos
- Department of Chemistry and Center for Behavioral Neuroscience, American University, Washington, D.C. 20016, United States
| | | | | | | | | | | |
Collapse
|
19
|
Abstract
Protein kinase C (PKC) isozymes belong to a family of Ser/Thr kinases whose activity is governed by reversible release of an autoinhibitory pseudosubstrate. For conventional and novel isozymes, this is effected by binding the lipid second messenger, diacylglycerol, but for atypical PKC isozymes, this is effected by binding protein scaffolds. PKC shot into the limelight following the discovery in the 1980s that the diacylglycerol-sensitive isozymes are "receptors" for the potent tumor-promoting phorbol esters. This set in place a concept that PKC isozymes are oncoproteins. Yet three decades of cancer clinical trials targeting PKC with inhibitors failed and, in some cases, worsened patient outcome. Emerging evidence from cancer-associated mutations and protein expression levels provide a reason: PKC isozymes generally function as tumor suppressors and their activity should be restored, not inhibited, in cancer therapies. And whereas not enough activity is associated with cancer, variants with enhanced activity are associated with degenerative diseases such as Alzheimer's disease. This review describes the tightly controlled mechanisms that ensure PKC activity is perfectly balanced and what happens when these controls are deregulated. PKC isozymes serve as a paradigm for the wisdom of Confucius: "to go beyond is as wrong as to fall short."
Collapse
Affiliation(s)
- Alexandra C Newton
- a Department of Pharmacology , University of California at San Diego , La Jolla , CA , USA
| |
Collapse
|
20
|
Abstract
There is a plethora of amphetamine derivatives exerting stimulant, euphoric, anti-fatigue, and hallucinogenic effects; all structural properties allowing these effects are contained within the amphetamine structure. In the first part of this review, the interaction of amphetamine with the dopamine transporter (DAT), crucially involved in its behavioral effects, is covered, as well as the role of dopamine synthesis, the vesicular monoamine transporter VMAT2, and organic cation 3 transporter (OCT3). The second part deals with requirements in amphetamine's effect on the kinases PKC, CaMKII, and ERK, whereas the third part focuses on where we are in developing anti-amphetamine therapeutics. Thus, treatments are discussed that target DAT, VMAT2, PKC, CaMKII, and OCT3. As is generally true for the development of therapeutics for substance use disorder, there are multiple preclinically promising specific compounds against (meth)amphetamine, for which further development and clinical trials are badly needed.
Collapse
Affiliation(s)
- Maarten E A Reith
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
| | - Margaret E Gnegy
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| |
Collapse
|
21
|
Carmona-Rosas G, Alcántara-Hernández R, Hernández-Espinosa DA. The role of β-arrestins in G protein-coupled receptor heterologous desensitization: A brief story. Methods Cell Biol 2018; 149:195-204. [PMID: 30616820 DOI: 10.1016/bs.mcb.2018.08.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
G protein-coupled receptors (GPCRs) are transmembrane proteins that have an important impact in a myriad of cellular functions. Posttranslational modifications on GPCRs are a key processes that allow these proteins to recruit other intracellular molecules. Among these modifications, phosphorylation is the most important way of desensitization of these receptors. Several research groups have described two different desensitization mechanisms: heterologous and homologous desensitization. The first one involves the phosphorylation of the receptors by protein kinases, such as PKC, following the desensitization and internalization of the receptor, while the second one involves the phosphorylation of the receptors by GRKs, allowing for the receptor to recruit β-arrestins to be desensitized and internalized. Interestingly, a few number of studies have described the participation of β-arrestins during the heterologous desensitization process. Hence, the aim of this review is to briefly explore the role that β-arrestins play during the heterologous desensitization of several GPCRs.
Collapse
|
22
|
Dąbrowska K, Albrecht J, Zielińska M. Protein kinase C-mediated impairment of glutamine outward transport and SN1 transporter distribution by ammonia in mouse cortical astrocytes. Neurochem Int 2018; 118:225-232. [DOI: 10.1016/j.neuint.2018.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 06/19/2018] [Accepted: 07/03/2018] [Indexed: 01/07/2023]
|
23
|
Tóth AD, Prokop S, Gyombolai P, Várnai P, Balla A, Gurevich VV, Hunyady L, Turu G. Heterologous phosphorylation-induced formation of a stability lock permits regulation of inactive receptors by β-arrestins. J Biol Chem 2018; 293:876-892. [PMID: 29146594 PMCID: PMC5777260 DOI: 10.1074/jbc.m117.813139] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 11/08/2017] [Indexed: 12/24/2022] Open
Abstract
β-Arrestins are key regulators and signal transducers of G protein-coupled receptors (GPCRs). The interaction between receptors and β-arrestins is generally believed to require both receptor activity and phosphorylation by GPCR kinases. In this study, we investigated whether β-arrestins are able to bind second messenger kinase-phosphorylated, but inactive receptors as well. Because heterologous phosphorylation is a common phenomenon among GPCRs, this mode of β-arrestin activation may represent a novel mechanism of signal transduction and receptor cross-talk. Here we demonstrate that activation of protein kinase C (PKC) by phorbol myristate acetate, Gq/11-coupled GPCR, or epidermal growth factor receptor stimulation promotes β-arrestin2 recruitment to unliganded AT1 angiotensin receptor (AT1R). We found that this interaction depends on the stability lock, a structure responsible for the sustained binding between GPCRs and β-arrestins, formed by phosphorylated serine-threonine clusters in the receptor's C terminus and two conserved phosphate-binding lysines in the β-arrestin2 N-domain. Using improved FlAsH-based serine-threonine clusters β-arrestin2 conformational biosensors, we also show that the stability lock not only stabilizes the receptor-β-arrestin interaction, but also governs the structural rearrangements within β-arrestins. Furthermore, we found that β-arrestin2 binds to PKC-phosphorylated AT1R in a distinct active conformation, which triggers MAPK recruitment and receptor internalization. Our results provide new insights into the activation of β-arrestins and reveal their novel role in receptor cross-talk.
Collapse
Affiliation(s)
- András D Tóth
- From the Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest H-1094, Hungary
| | - Susanne Prokop
- From the Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest H-1094, Hungary
| | - Pál Gyombolai
- From the Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest H-1094, Hungary
- the MTA-SE Laboratory of Molecular Physiology, Hungarian Academy of Sciences and Semmelweis University, Budapest H-1094, Hungary, and
| | - Péter Várnai
- From the Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest H-1094, Hungary
- the MTA-SE Laboratory of Molecular Physiology, Hungarian Academy of Sciences and Semmelweis University, Budapest H-1094, Hungary, and
| | - András Balla
- From the Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest H-1094, Hungary
- the MTA-SE Laboratory of Molecular Physiology, Hungarian Academy of Sciences and Semmelweis University, Budapest H-1094, Hungary, and
| | - Vsevolod V Gurevich
- the Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| | - László Hunyady
- From the Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest H-1094, Hungary,
- the MTA-SE Laboratory of Molecular Physiology, Hungarian Academy of Sciences and Semmelweis University, Budapest H-1094, Hungary, and
| | - Gábor Turu
- From the Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest H-1094, Hungary
- the MTA-SE Laboratory of Molecular Physiology, Hungarian Academy of Sciences and Semmelweis University, Budapest H-1094, Hungary, and
| |
Collapse
|
24
|
Watson U, Jain R, Asthana S, Saini DK. Spatiotemporal Modulation of ERK Activation by GPCRs. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 338:111-140. [DOI: 10.1016/bs.ircmb.2018.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
|
25
|
Visualization of ligand-induced dopamine D 2S and D 2L receptor internalization by TIRF microscopy. Sci Rep 2017; 7:10894. [PMID: 28883522 PMCID: PMC5589927 DOI: 10.1038/s41598-017-11436-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 08/23/2017] [Indexed: 01/11/2023] Open
Abstract
G protein-coupled receptors (GPCRs), including the dopamine receptors, represent a group of important pharmacological targets. Upon agonist binding, GPCRs frequently undergo internalization, a process that is known to attenuate functional responses upon prolonged exposure to agonists. In this study, internalization was visualized by means of total internal reflection fluorescence (TIRF) microscopy at a level of discrete single events near the plasma membrane with high spatial resolution. A novel method has been developed to determine the relative extent of internalized fluorescent receptor-ligand complexes by comparative fluorescence quantification in living CHO cells. The procedure entails treatment with the reducing agent sodium borohydride, which converts cyanine-based fluorescent ligands on the membrane surface to a long-lived reduced form. Because the highly polar reducing agent is not able to pass the cell membrane, the fluorescent receptor-ligand complexes located in internalized compartments remain fluorescent under TIRF illumination. We applied the method to investigate differences of the short (D2S) and the long (D2L) isoforms of dopamine D2 receptors in their ability to undergo agonist-induced internalization.
Collapse
|
26
|
Yang Z, Yang F, Zhang D, Liu Z, Lin A, Liu C, Xiao P, Yu X, Sun JP. Phosphorylation of G Protein-Coupled Receptors: From the Barcode Hypothesis to the Flute Model. Mol Pharmacol 2017; 92:201-210. [PMID: 28246190 DOI: 10.1124/mol.116.107839] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 02/23/2017] [Indexed: 02/14/2025] Open
Abstract
Seven transmembrane G protein-coupled receptors (GPCRs) are often phosphorylated at the C terminus and on intracellular loops in response to various extracellular stimuli. Phosphorylation of GPCRs by GPCR kinases and certain other kinases can promote the recruitment of arrestin molecules. The arrestins critically regulate GPCR functions not only by mediating receptor desensitization and internalization, but also by redirecting signaling to G protein-independent pathways via interactions with numerous downstream effector molecules. Accumulating evidence over the past decade has given rise to the phospho-barcode hypothesis, which states that ligand-specific phosphorylation patterns of a receptor direct its distinct functional outcomes. Our recent work using unnatural amino acid incorporation and fluorine-19 nuclear magnetic resonance (19F-NMR) spectroscopy led to the flute model, which provides preliminary insight into the receptor phospho-coding mechanism, by which receptor phosphorylation patterns are recognized by an array of phosphate-binding pockets on arrestin and are translated into distinct conformations. These selective conformations are recognized by various effector molecules downstream of arrestin. The phospho-barcoding mechanism enables arrestin to recognize a wide range of phosphorylation patterns of GPCRs, contributing to their diverse functions.
Collapse
Affiliation(s)
- Zhao Yang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People's Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People's Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.)
| | - Fan Yang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People's Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People's Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.)
| | - Daolai Zhang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People's Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People's Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.)
| | - Zhixin Liu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People's Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People's Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.)
| | - Amy Lin
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People's Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People's Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.)
| | - Chuan Liu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People's Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People's Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.)
| | - Peng Xiao
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People's Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People's Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.)
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People's Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People's Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.)
| | - Jin-Peng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology (Z.Y., Z.L., C.L., P.X., J.-P.S.), Department of Physiology (F.Y., X.Y.), Shandong University School of Medicine, Jinan, Shandong, People's Republic of China; School of Pharmacy, Binzhou Medical University, Yantai, Shandong, People's Republic of China (D.Z.); School of Medicine, Duke University, Durham, North Carolina (A.L., J.-P.S.)
| |
Collapse
|
27
|
Okamoto Y, Shikano S. Differential phosphorylation signals control endocytosis of GPR15. Mol Biol Cell 2017; 28:2267-2281. [PMID: 28615320 PMCID: PMC5555655 DOI: 10.1091/mbc.e16-09-0627] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 06/05/2017] [Accepted: 06/07/2017] [Indexed: 12/30/2022] Open
Abstract
GPR15 undergoes a ligand-independent endocytosis, which requires phosphorylation of a distal C-terminal Ser-357 mediated by multiple basophilic kinases. The functional role of Ser-357 in endocytosis is distinct from that of a conserved Ser/Thr cluster, which is more responsible for the use of GRKs and β-arrestin. GPR15 is an orphan G protein–coupled receptor (GPCR) that serves for an HIV coreceptor and was also recently found as a novel homing receptor for T-cells implicated in colitis. We show that GPR15 undergoes a constitutive endocytosis in the absence of ligand. The endocytosis was clathrin dependent and partially dependent on β-arrestin in HEK293 cells, and nearly half of the internalized GPR15 receptors were recycled to the plasma membrane. An Ala mutation of the distal C-terminal Arg-354 or Ser-357, which forms a consensus phosphorylation site for basophilic kinases, markedly reduced the endocytosis, whereas phosphomimetic mutation of Ser-357 to Asp did not. Ser-357 was phosphorylated in vitro by multiple kinases, including PKA and PKC, and pharmacological activation of these kinases enhanced both phosphorylation of Ser-357 and endocytosis of GPR15. These results suggested that Ser-357 phosphorylation critically controls the ligand-independent endocytosis of GPR15. The functional role of Ser-357 in endocytosis was distinct from that of a conserved Ser/Thr cluster in the more proximal C-terminus, which was responsible for the β-arrestin– and GPCR kinase–dependent endocytosis of GPR15. Thus phosphorylation signals may differentially control cell surface density of GPR15 through endocytosis.
Collapse
Affiliation(s)
- Yukari Okamoto
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607-7170
| | - Sojin Shikano
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607-7170
| |
Collapse
|
28
|
Protein kinase C as a tumor suppressor. Semin Cancer Biol 2017; 48:18-26. [PMID: 28476658 DOI: 10.1016/j.semcancer.2017.04.017] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 03/31/2017] [Accepted: 04/28/2017] [Indexed: 01/01/2023]
Abstract
Protein kinase C (PKC) has historically been considered an oncoprotein. This stems in large part from the discovery in the early 1980s that PKC is directly activated by tumor-promoting phorbol esters. Yet three decades of clinical trials using PKC inhibitors in cancer therapies not only failed, but in some cases worsened patient outcome. Why has targeting PKC in cancer eluded successful therapies? Recent studies looking at the disease for insight provide an explanation: cancer-associated mutations in PKC are generally loss-of-function (LOF), supporting an unexpected function as tumor suppressors. And, contrasting with LOF mutations in cancer, germline mutations that enhance the activity of some PKC isozymes are associated with degenerative diseases such as Alzheimer's disease. This review provides a background on the diverse mechanisms that ensure PKC is only active when, where, and for the appropriate duration needed and summarizes recent findings converging on a paradigm reversal: PKC family members generally function by suppressing, rather than promoting, survival signaling.
Collapse
|
29
|
Mikelman S, Mardirossian N, Gnegy ME. Tamoxifen and amphetamine abuse: Are there therapeutic possibilities? J Chem Neuroanat 2016; 83-84:50-58. [PMID: 27585851 DOI: 10.1016/j.jchemneu.2016.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 08/05/2016] [Accepted: 08/14/2016] [Indexed: 12/11/2022]
Abstract
Although best known as a selective estrogen receptor modulator (SERM), tamoxifen is a drug with a wide range of activities. Tamoxifen has demonstrated some efficacy has a therapeutic for bipolar mania and is believed to exert these effects through inhibition of protein kinase C (PKC). As the symptoms of amphetamine treatment in rodents are believed to mimic the symptoms of a manic episode, many of the preclinical studies for this indication have demonstrated that tamoxifen inhibits amphetamine action. The amphetamine-induced increase in extracellular dopamine which gives rise to the 'manic' effects is due to interaction of amphetamine with the dopamine transporter. We and others have demonstrated that PKC reduces amphetamine-induced reverse transport through the dopamine transporter. In this review, we will outline the actions of tamoxifen as a SERM and further detail another known action of tamoxifen-inhibition of PKC. We will summarize the literature showing how tamoxifen affects amphetamine action. Finally, we will present our hypothesis that tamoxifen, or an analog, could be used therapeutically to reduce amphetamine abuse in addition to treating mania.
Collapse
Affiliation(s)
- Sarah Mikelman
- Department of Pharmacology, 2220E MSRB III, 1150 West Medical Center Drive, University of Michigan Medical School, Ann Arbor, MI 28109-5632, United States
| | - Natalie Mardirossian
- Department of Pharmacology, 2220E MSRB III, 1150 West Medical Center Drive, University of Michigan Medical School, Ann Arbor, MI 28109-5632, United States
| | - Margaret E Gnegy
- Department of Pharmacology, 2220E MSRB III, 1150 West Medical Center Drive, University of Michigan Medical School, Ann Arbor, MI 28109-5632, United States.
| |
Collapse
|
30
|
Heterologous, PKC-Mediated Desensitization of Human Histamine H3 Receptors Expressed in CHO-K1 Cells. Neurochem Res 2016; 41:2415-24. [PMID: 27350581 DOI: 10.1007/s11064-016-1954-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 05/06/2016] [Accepted: 05/11/2016] [Indexed: 12/11/2022]
Abstract
Desensitization is a major mechanism to regulate the functional response of G protein-coupled receptors. In this work we studied whether the human histamine H3 receptor of 445 amino acids (hH3R445) experiences heterologous desensitization mediated by PKC activation. Bioinformatic analysis indicated the presence of Serine and Threonine residues susceptible of PKC-mediated phosphorylation on the third intracellular loop and the carboxyl terminus of the hH3R445. In CHO-K1 cells stably transfected with the hH3R445 direct PKC activation by phorbol 12-myristate 13-acetate (TPA, 200 nM) abolished H3R-mediated inhibition of forskolin-stimulated cAMP accumulation. Activation of endogenous purinergic receptors by ATP (adenosine 5'-triphosphate, 10 μM) increased the free calcium intracellular concentration ([Ca(2+)]i) confirming their coupling to phospholipase C stimulation. Incubation with ATP also abolished H3R-mediated inhibition of forskolin-induced cAMP accumulation, and this effect was prevented by the PKC inhibitors Ro-31-8220 and Gö-6976. Pre-incubation with TPA or ATP reduced H3R-mediated stimulation of [(35)S]-GTPγS binding to membranes from CHO-K1-hH3R445 cells by 39.7 and 54.2 %, respectively, with no change in the agonist potency, and the effect was prevented by either Ro-31-8220 or Gö-6976. Exposure to ATP or TPA also resulted in the loss of cell surface H3Rs (-30.4 and -45.1 %) as evaluated by [(3)H]-NMHA binding to intact cells. These results indicate that the hH3R445 undergoes heterologous desensitization upon activation of receptors coupled to PKC stimulation.
Collapse
|
31
|
Zestos AG, Mikelman SR, Kennedy RT, Gnegy ME. PKCβ Inhibitors Attenuate Amphetamine-Stimulated Dopamine Efflux. ACS Chem Neurosci 2016; 7:757-66. [PMID: 26996926 DOI: 10.1021/acschemneuro.6b00028] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Amphetamine abuse afflicts over 13 million people, and there is currently no universally accepted treatment for amphetamine addiction. Amphetamine serves as a substrate for the dopamine transporter and reverses the transporter to cause an increase in extracellular dopamine. Activation of the beta subunit of protein kinase C (PKCβ) enhances extracellular dopamine in the presence of amphetamine by facilitating the reverse transport of dopamine and internalizing the D2 autoreceptor. We previously demonstrated that PKCβ inhibitors block amphetamine-stimulated dopamine efflux in synaptosomes from rat striatum in vitro. In this study, we utilized in vivo microdialysis in live, behaving rats to assess the effect of the PKCβ inhibitors, enzastaurin and ruboxistaurin, on amphetamine-stimulated locomotion and increases in monoamines and their metabolites. A 30 min perfusion of the nucleus accumbens core with 1 μM enzastaurin or 1 μM ruboxistaurin reduced efflux of dopamine and its metabolite 3-methoxytyramine induced by amphetamine by approximately 50%. The inhibitors also significantly reduced amphetamine-stimulated extracellular levels of norepinephrine. The stimulation of locomotor behavior by amphetamine, measured simultaneously with the analytes, was comparably reduced by the PKCβ inhibitors. Using a stable isotope label retrodialysis procedure, we determined that ruboxistaurin had no effect on basal levels of dopamine, norepinephrine, glutamate, or GABA. In addition, normal uptake function through the dopamine transporter was unaltered by the PKCβ inhibitors, as measured in rat synaptosomes. Our results support the utility of using PKCβ inhibitors to reduce the effects of amphetamine.
Collapse
Affiliation(s)
- Alexander G. Zestos
- Department
of Pharmacology, University of Michigan, 2301 MSRB III, 1150 W. Medical Center
Drive, Ann Arbor, Michigan 48109-5632, United States
- Department
of Chemistry, University of Michigan, 9300 North University Avenue, Ann Arbor, Michigan 48105, United States
| | - Sarah R. Mikelman
- Department
of Pharmacology, University of Michigan, 2301 MSRB III, 1150 W. Medical Center
Drive, Ann Arbor, Michigan 48109-5632, United States
| | - Robert T. Kennedy
- Department
of Pharmacology, University of Michigan, 2301 MSRB III, 1150 W. Medical Center
Drive, Ann Arbor, Michigan 48109-5632, United States
- Department
of Chemistry, University of Michigan, 9300 North University Avenue, Ann Arbor, Michigan 48105, United States
| | - Margaret E. Gnegy
- Department
of Pharmacology, University of Michigan, 2301 MSRB III, 1150 W. Medical Center
Drive, Ann Arbor, Michigan 48109-5632, United States
| |
Collapse
|
32
|
Hunter MR, Grimsey NL, Glass M. Sulfation of the FLAG epitope is affected by co-expression of G protein-coupled receptors in a mammalian cell model. Sci Rep 2016; 6:27316. [PMID: 27273047 PMCID: PMC4895180 DOI: 10.1038/srep27316] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 05/15/2016] [Indexed: 01/13/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are important therapeutic targets and therefore extensively studied. Like most transmembrane proteins, there has been considerable difficulty in developing reliable specific antibodies for them. To overcome this, epitope tags are often used to facilitate antibody recognition in studies on fundamental receptor signalling and trafficking. In our study of cannabinoid CB1/dopamine D2 interactions we sought to generate HEK293 cells expressing FLAG-tagged D2 for use in antibody-based assays of GPCR localisation and trafficking activity, however observed that stable FLAG-hD2 expression was particularly challenging to maintain. In contrast, when expressed in cell lines expressing hCB1 robust and stable FLAG-hD2 expression was observed. We hypothesised that co-expression of CB1 might stabilise surface FLAG-hD2 expression, and therefore investigated this further. Here, we describe the observation that co-expression of either cannabinoid CB1 or CB2 receptors in HEK293 decreases the sulfation of a FLAG epitope appended at the N-terminus of the dopamine D2 receptor. Sulfation alters epitope recognition by some anti-FLAG antibodies, leading to the detection of fewer receptors, even though expression is maintained. This demonstrates that cannabinoid receptor expression modifies posttranslational processing of the FLAG-hD2 receptor, and importantly, has wider implications for the utilisation and interpretation of receptor studies involving epitope tags.
Collapse
Affiliation(s)
- Morag Rose Hunter
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - Natasha Lillia Grimsey
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| | - Michelle Glass
- Department of Pharmacology and Clinical Pharmacology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
33
|
Alternative Splicing in Ca(V)2.2 Regulates Neuronal Trafficking via Adaptor Protein Complex-1 Adaptor Protein Motifs. J Neurosci 2016; 35:14636-52. [PMID: 26511252 DOI: 10.1523/jneurosci.3034-15.2015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
N-type voltage-gated calcium (Ca(V)2.2) channels are expressed in neurons and targeted to the plasma membrane of presynaptic terminals, facilitating neurotransmitter release. Here, we find that the adaptor protein complex-1 (AP-1) mediates trafficking of Ca(V)2.2 from the trans-Golgi network to the cell surface. Examination of splice variants of Ca(V)2.2, containing either exon 37a (selectively expressed in nociceptors) or 37b in the proximal C terminus, reveal that canonical AP-1 binding motifs, YxxΦ and [DE]xxxL[LI], present only in exon 37a, enhance intracellular trafficking of exon 37a-containing Ca(V)2.2 to the axons and plasma membrane of rat DRG neurons. Finally, we identify differential effects of dopamine-2 receptor (D2R) and its agonist-induced activation on trafficking of Ca(V)2.2 isoforms. D2R slowed the endocytosis of Ca(V)2.2 containing exon 37b, but not exon 37a, and activation by the agonist quinpirole reversed the effect of the D2R. Our work thus reveals key mechanisms involved in the trafficking of N-type calcium channels.
Collapse
|
34
|
Schmieg N, Rocchi C, Romeo S, Maggio R, Millan MJ, Mannoury la Cour C. Dysbindin-1 modifies signaling and cellular localization of recombinant, human D₃ and D₂ receptors. J Neurochem 2016; 136:1037-51. [PMID: 26685100 DOI: 10.1111/jnc.13501] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 12/09/2015] [Indexed: 01/20/2023]
Abstract
Dystrobrevin binding protein-1 (dysbindin-1), a candidate gene for schizophrenia, modulates cognition, synaptic plasticity and frontocortical circuitry and interacts with glutamatergic and dopaminergic transmission. Loss of dysbindin-1 modifies cellular trafficking of dopamine (DA) D2 receptors to increase cell surface expression, but its influence upon signaling has never been characterized. Further, the effects of dysbindin-1 upon closely related D3 receptors remain unexplored. Hence, we examined the impact of dysbindin-1 (isoform A) co-expression on the localization and coupling of human D2L and D3 receptors stably expressed in Chinese hamster ovary or SH-SY5Y cells lacking endogenous dysbindin-1. Dysbindin-1 co-transfection decreased cell surface expression of both D3 and D2L receptors. Further, while their affinity for DA was unchanged, dysbindin-1 reduced the magnitude and potency of DA-induced adenylate cylase recruitment/cAMP production. Dysbindin-1 also blunted the amplitude of DA-induced phosphorylation of ERK1/2 and Akt at both D2L and D3 receptors without, in contrast to cAMP, affecting the potency of DA. Interference with calveolin/clathrin-mediated processes of internalization prevented the modification by dysbindin-1 of ERK1/2 and adenylyl cyclase stimulation at D2L and D3 receptors. Finally, underpinning the specificity of the influence of dysbindin-1 on D2L and D3 receptors, dysbindin-1 did not modify recruitment of adenylyl cyclase by D1 receptors. These observations demonstrate that dysbindin-1 influences cell surface expression of D3 in addition to D2L receptors, and that it modulates activation of their signaling pathways. Accordingly, both a deficiency and an excess of dysbindin-1 may be disruptive for dopaminergic transmission, supporting its link to schizophrenia and other CNS disorders. Dysbindin-1, a candidate gene for schizophrenia, alters D2 receptors cell surface expression. We demonstrate that dysbindin-1 expression also influences cell surface levels of D3 receptors. Further, Dysbindin-1 reduces DA-induced adenylate cylase recruitment/cAMP production and modifies major signaling pathways (Akt and extracellular signal-regulated kinases1/2 (ERK1/2)) of both D2 and D3 receptors. Dysbindin-1 modulates thus D2 and D3 receptor signaling, supporting a link to schizophrenia.
Collapse
Affiliation(s)
- Nathalie Schmieg
- PIT-Neuropsychiatry, Institut de Recherches Servier, Centre de Recherches de Croissy, Croissy-sur-Seine, France
| | - Cristina Rocchi
- Biotechnological and Applied Clinical Sciences Department, University of L'Aquila, L'Aquila, Italy
| | - Stefania Romeo
- Biotechnological and Applied Clinical Sciences Department, University of L'Aquila, L'Aquila, Italy
| | - Roberto Maggio
- Biotechnological and Applied Clinical Sciences Department, University of L'Aquila, L'Aquila, Italy
| | - Mark J Millan
- PIT-Neuropsychiatry, Institut de Recherches Servier, Centre de Recherches de Croissy, Croissy-sur-Seine, France
| | - Clotilde Mannoury la Cour
- PIT-Neuropsychiatry, Institut de Recherches Servier, Centre de Recherches de Croissy, Croissy-sur-Seine, France
| |
Collapse
|
35
|
Zheng M, Zhang X, Guo S, Zhang X, Choi HJ, Lee MY, Kim KM. PKCβII inhibits the ubiquitination of β-arrestin2 in an autophosphorylation-dependent manner. FEBS Lett 2015; 589:3929-37. [DOI: 10.1016/j.febslet.2015.10.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 09/07/2015] [Accepted: 10/23/2015] [Indexed: 01/19/2023]
|
36
|
Matsuoka H, Inoue M. Src mediates endocytosis of TWIK-related acid-sensitive K+ 1 channels in PC12 cells in response to nerve growth factor. Am J Physiol Cell Physiol 2015; 309:C251-63. [DOI: 10.1152/ajpcell.00354.2014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 06/10/2015] [Indexed: 01/17/2023]
Abstract
TWIK-related acid-sensitive K+ (TASK) channels produce background K+ currents. We elucidated that TASK1 channels in rat adrenal medullary cells and PC12 cells are internalized in a clathrin-dependent manner in response to nerve growth factor (NGF). Here, the molecular mechanism for this internalization in PC12 cells was explored. The combination of enzyme inhibitors with tropomyosin receptor kinase A mutants revealed that the internalization was mediated by both phospholipase C and phosphatidylinositol 3-kinase pathways that converge on protein kinase C with the consequent activation of Src, a nonreceptor tyrosine kinase. The NGF-induced endocytosis of TASK1 channels did not occur in the presence of the Src inhibitor or with the expression of a kinase-dead Src mutant. Additionally, NGF induced a transient colocalization of Src with the TASK1 channel, but not the TASK1 mutant, in which tyrosine at 370 was replaced with phenylalanine. This TASK1 mutant showed no increase in tyrosine phosphorylation and markedly diminished internalization in response to NGF. We concluded that NGF induces endocytosis of TASK1 channels via tyrosine phosphorylation in its carboxyl terminus.
Collapse
Affiliation(s)
- Hidetada Matsuoka
- Department of Cell and Systems Physiology, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan
| | - Masumi Inoue
- Department of Cell and Systems Physiology, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan
| |
Collapse
|
37
|
Luderman KD, Chen R, Ferris MJ, Jones SR, Gnegy ME. Protein kinase C beta regulates the D₂-like dopamine autoreceptor. Neuropharmacology 2015; 89:335-41. [PMID: 25446677 DOI: 10.1016/j.neuropharm.2014.10.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 10/03/2014] [Accepted: 10/07/2014] [Indexed: 01/15/2023]
Abstract
The focus of this study was the regulation of the D2-like dopamine autoreceptor (D2 autoreceptor) by protein kinase Cβ, a member of the protein kinase C (PKC) family. Together with the dopamine transporter, the D2 autoreceptor regulates the level of extracellular dopamine and thus dopaminergic signaling. PKC regulates neuronal signaling via several mechanisms, including desensitizing autoreceptors to increase the release of several different neurotransmitters. Here, using both PKCβ(-/-) mice and specific PKCβ inhibitors, we demonstrated that a lack of PKCβ activity enhanced the D2 autoreceptor-stimulated decrease in dopamine release following both chemical and electrical stimulations. Inhibition of PKCβ increased surface localization of D2R in mouse striatal synaptosomes, which could underlie the greater sensitivity to quinpirole following inhibition of PKCβ. PKCβ(-/-) mice displayed greater sensitivity to the quinpirole-induced suppression of locomotor activity, demonstrating that the regulation of the D2 autoreceptor by PKCβ is physiologically significant. Overall, we have found that PKCβ downregulates the D2 autoreceptor, providing an additional layer of regulation for dopaminergic signaling. We propose that in the absence of PKCβ activity, surface D2 autoreceptor localization and thus D2 autoreceptor signaling is increased, leading to less dopamine in the extracellular space and attenuated dopaminergic signaling.
Collapse
Affiliation(s)
- Kathryn D Luderman
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5632, USA.
| | | | | | | | | |
Collapse
|
38
|
Deming JD, Shin JA, Lim K, Lee EJ, Van Craenenbroeck K, Craft CM. Dopamine receptor D4 internalization requires a beta-arrestin and a visual arrestin. Cell Signal 2015; 27:2002-13. [PMID: 26169958 DOI: 10.1016/j.cellsig.2015.06.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 06/30/2015] [Indexed: 11/25/2022]
Abstract
PURPOSE The G-protein coupled receptor (GPCR) Dopamine Receptor D4 (DRD4) plays an essential role in cAMP regulation and gap junctional coupling in the photoreceptors, where DRD4 expression is under circadian control. Previous in vitro transfection studies of human DRD4 desensitization have reported that DRD4 is not internalized upon dopamine stimulation when beta-arrestin is co-transfected with DRD4. We hypothesized that the visual arrestins, ARR1 and ARR4, play a modulatory role in DRD4 desensitization in the photoreceptors. METHODS To test this hypothesis, immunohistochemistry analysis of mouse retinas was used to determine the cellular localization of beta-arrestins and DRD4 in photoreceptors. In vitro studies were performed in HEK293T cells transiently transfected with human DRD4 and arrestins. First, co-immunoprecipitation experiments were executed to test protein-protein interactions and to investigate the effect of dopamine stimulation. Second, immunohistochemistry analysis was implemented to study DRD4 internalization and translocation of ARR4. RESULTS Immunohistochemistry studies of mouse retinas confirmed the expression of beta-arrestin 2, ARR1 and ARR4, as well as DRD4 in mouse cone photoreceptor inner segments. Co-immunoprecipitation experiments revealed a dopamine-dependent protein-protein interaction between human DRD4 and ARR4. In vitro internalization experiments showed that no detectable internalization of DRD4 was observed with any single arrestin co-transfected. However, a dopamine-dependent internalization of DRD4 was observed with three out of six sets of two arrestins co-transfected with DRD4. Each of these pairs of arrestins contained one visual arrestin and one beta-arrestin, and no internalization was observed with either two visual arrestins or two beta-arrestins. Additional time-course experiments revealed that in vitro, ARR4 translocates to co-localize with DRD4 at the plasma membrane in response to 30min of dopamine stimulation. CONCLUSIONS The results have functional implications and we hypothesize that the desensitization and internalization of DRD4 in photoreceptors are synergistically mediated by both visual and beta-arrestins. These results are additionally unique because they demonstrate for the first time that at least one G-protein coupled receptor, DRD4, requires two arrestins for desensitization and internalization, and opens up the possibility that other G-protein coupled receptors may require more than one arrestin for desensitization and/or internalization.
Collapse
Affiliation(s)
- Janise D Deming
- Mary D. Allen Laboratory for Vision Research, USC Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, 2250 Alcazar Street, Clinical Science Annex 215, Los Angeles, CA 90033, USA.
| | - Jung-A Shin
- Mary D. Allen Laboratory for Vision Research, USC Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, 2250 Alcazar Street, Clinical Science Annex 215, Los Angeles, CA 90033, USA; Department of Anatomy, School of Medicine, Ewha Womans University, Seoul 158-710, Republic of Korea.
| | - Kayleen Lim
- Mary D. Allen Laboratory for Vision Research, USC Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, 2250 Alcazar Street, Clinical Science Annex 215, Los Angeles, CA 90033, USA.
| | - Eun-Jin Lee
- Mary D. Allen Laboratory for Vision Research, USC Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, 2250 Alcazar Street, Clinical Science Annex 215, Los Angeles, CA 90033, USA; Department of Biomedical Engineering, University of Southern California Viterbi School of Engineering, 1042 Downey Way, Los Angeles, CA 90089-1111, USA.
| | - Kathleen Van Craenenbroeck
- Laboratory of GPCR Expression and Signal Transduction (L-GEST), Ghent University-UGent, K.L. Ledeganckstraat 35, B-9000 Gent, Belgium.
| | - Cheryl Mae Craft
- Mary D. Allen Laboratory for Vision Research, USC Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, 2250 Alcazar Street, Clinical Science Annex 215, Los Angeles, CA 90033, USA; Department of Cell & Neurobiology, Keck School of Medicine of the University of Southern California, 2250 Alcazar Street, Clinical Science Center 135H, Los Angeles, CA 90033, USA.
| |
Collapse
|
39
|
Octeau JC, Schrader JM, Masuho I, Sharma M, Aiudi C, Chen CK, Kovoor A, Celver J. G protein beta 5 is targeted to D2-dopamine receptor-containing biochemical compartments and blocks dopamine-dependent receptor internalization. PLoS One 2014; 9:e105791. [PMID: 25162404 PMCID: PMC4146516 DOI: 10.1371/journal.pone.0105791] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 07/23/2014] [Indexed: 11/18/2022] Open
Abstract
G beta 5 (Gbeta5, Gβ5) is a unique G protein β subunit that is thought to be expressed as an obligate heterodimer with R7 regulator of G protein signaling (RGS) proteins instead of with G gamma (Gγ) subunits. We found that D2-dopamine receptor (D2R) coexpression enhances the expression of Gβ5, but not that of the G beta 1 (Gβ1) subunit, in HEK293 cells, and that the enhancement of expression occurs through a stabilization of Gβ5 protein. We had previously demonstrated that the vast majority of D2R either expressed endogenously in the brain or exogenously in cell lines segregates into detergent-resistant biochemical fractions. We report that when expressed alone in HEK293 cells, Gβ5 is highly soluble, but is retargeted to the detergent-resistant fraction after D2R coexpression. Furthermore, an in-cell biotin transfer proximity assay indicated that D2R and Gβ5 segregating into the detergent-resistant fraction specifically interacted in intact living cell membranes. Dopamine-induced D2R internalization was blocked by coexpression of Gβ5, but not Gβ1. However, the same Gβ5 coexpression levels had no effect on agonist-induced internalization of the mu opioid receptor (MOR), cell surface D2R levels, dopamine-mediated recruitment of β-arrestin to D2R, the amplitude of D2R-G protein coupling, or the deactivation kinetics of D2R-activated G protein signals. The latter data suggest that the interactions between D2R and Gβ5 are not mediated by endogenously expressed R7 RGS proteins.
Collapse
Affiliation(s)
- J. Christopher Octeau
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, United States of America
| | - Joseph M. Schrader
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, United States of America
| | - Ikuo Masuho
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, United States of America
| | - Meenakshi Sharma
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, United States of America
| | - Christopher Aiudi
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, United States of America
| | - Ching-Kang Chen
- Cullen Eye Institute, Baylor College of Medicine, Houston, Texas, United States of America
| | - Abraham Kovoor
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, United States of America
- * E-mail: (AK); (JC)
| | - Jeremy Celver
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island, United States of America
- * E-mail: (AK); (JC)
| |
Collapse
|
40
|
Koyama S, Mori M, Kanamaru S, Sazawa T, Miyazaki A, Terai H, Hirose S. Obesity attenuates D2 autoreceptor-mediated inhibition of putative ventral tegmental area dopaminergic neurons. Physiol Rep 2014; 2:e12004. [PMID: 24793981 PMCID: PMC4098733 DOI: 10.14814/phy2.12004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The ventral tegmental area (VTA) in the midbrain is important for food reward. High‐fat containing palatable foods have reinforcing effects and accelerate obesity. We have previously reported that diet‐induced obesity selectively decreased the spontaneous activity of VTA GABA neurons, but not dopamine neurons. The spontaneous activity of VTA dopamine neurons is regulated by D2 autoreceptors. In this study, we hypothesized that obesity would affect the excitability of VTA dopamine neurons via D2 autoreceptors. To examine this hypothesis, we compared D2 receptor‐mediated responses of VTA dopamine neurons between lean and obese mice. Mice fed on a high‐fat (45%) diet and mice fed on a standard diet were used as obese and lean models, respectively. Brain slice preparations were made from these two groups. Spontaneous activity of VTA neurons was recorded by extracellular recording. Putative VTA dopamine neurons were identified by firing inhibition with a D2 receptor agonist quinpirole, and electrophysiological criteria (firing frequency <5 Hz and action potential current duration >1.2 msec). Single‐dose application of quinpirole (3−100 nmol/L) exhibited similar firing inhibition of putative VTA dopamine neurons between lean and obese mice. In stepwise application by increasing quinpirole concentrations of 3, 10, 30, and 100 nmol/L subsequently, quinpirole‐induced inhibition of firing decreased in putative VTA dopamine neurons of obese mice compared with those of lean mice. In conclusion, high‐fat diet‐induced obesity attenuated D2 receptor‐mediated inhibition of putative VTA dopamine neurons due to the acceleration of D2 receptor desensitization. High‐fat diet‐induced obesity did not affect firing properties of putative ventral tegmental area (VTA) dopamine neurons. However, obesity attenuated dopamine D2 receptor‐mediated inhibition of putative VTA dopamine neurons due to the acceleration of receptor desensitization. In obesity, decrease in D2 receptor‐mediated autoinhibition may contribute to pathophysiology of reinforcing process in feeding.
Collapse
Affiliation(s)
- Susumu Koyama
- Department of Psychosomatic Medicine, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
41
|
Nimitvilai S, Herman M, You C, Arora DS, McElvain MA, Roberto M, Brodie MS. Dopamine D2 receptor desensitization by dopamine or corticotropin releasing factor in ventral tegmental area neurons is associated with increased glutamate release. Neuropharmacology 2014; 82:28-40. [PMID: 24657149 DOI: 10.1016/j.neuropharm.2014.03.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 03/06/2014] [Accepted: 03/10/2014] [Indexed: 10/25/2022]
Abstract
Neurons of the ventral tegmental area (VTA) are the source of dopaminergic (DAergic) input to important brain regions related to addiction. Prolonged exposure of these VTA neurons to moderate concentrations of dopamine (DA) causes a time-dependent decrease in DA-induced inhibition, a complex desensitization called DA inhibition reversal (DIR). DIR is mediated by conventional protein kinase C (cPKC) through concurrent stimulation of D2 and D1-like DA receptors, or by D2 stimulation concurrent with activation of some Gq-linked receptors. Corticotropin releasing factor (CRF) acts via Gq, and can modulate glutamater neurotransmission in the VTA. In the present study, we used brain slice electrophysiology to characterize the interaction of DA, glutamate antagonists, and CRF agonists in the induction and maintenance of DIR in the VTA. Glutamate receptor antagonists blocked induction but not maintenance of DIR. Putative blockers of neurotransmitter release and store-operated calcium channels blocked and reversed DIR. CRF and the CRF agonist urocortin reversed inhibition produced by the D2 agonist quinpirole, consistent with our earlier work indicating that Gq activation reverses quinpirole-mediated inhibition. In whole cell recordings, the combination of urocortin and quinpirole, but not either agent alone, increased spontaneous excitatory postsynaptic currents (sEPSCs) in VTA neurons. Likewise, the combination of a D1-like receptor agonist and quinpirole, but not either agent alone, increased sEPSCs in VTA neurons. In summary, desensitization of D2 receptors induced by dopamine or CRF on DAergic VTA neurons is associated with increased glutamatergic signaling in the VTA.
Collapse
Affiliation(s)
- Sudarat Nimitvilai
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 S. Wolcott, Room E-202, M/C 901, Chicago, IL 60612-7342, USA
| | - Melissa Herman
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, SP30-1150, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Chang You
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 S. Wolcott, Room E-202, M/C 901, Chicago, IL 60612-7342, USA
| | - Devinder S Arora
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 S. Wolcott, Room E-202, M/C 901, Chicago, IL 60612-7342, USA
| | - Maureen A McElvain
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 S. Wolcott, Room E-202, M/C 901, Chicago, IL 60612-7342, USA
| | - Marisa Roberto
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, SP30-1150, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Mark S Brodie
- Department of Physiology and Biophysics, University of Illinois at Chicago, 835 S. Wolcott, Room E-202, M/C 901, Chicago, IL 60612-7342, USA.
| |
Collapse
|
42
|
Burns RN, Singh M, Senatorov IS, Moniri NH. Mechanisms of homologous and heterologous phosphorylation of FFA receptor 4 (GPR120): GRK6 and PKC mediate phosphorylation of Thr³⁴⁷, Ser³⁵⁰, and Ser³⁵⁷ in the C-terminal tail. Biochem Pharmacol 2014; 87:650-9. [PMID: 24412271 DOI: 10.1016/j.bcp.2013.12.016] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 12/26/2013] [Accepted: 12/27/2013] [Indexed: 01/26/2023]
Abstract
Free fatty acid receptor 4 (FFA4), previously known as GPR120, is a G protein-coupled receptor that promotes numerous anti-inflammatory and antidiabetic effects upon its agonism by long chained unsaturated fatty acids. We have previously demonstrated that agonism of FFA4 with docosahexaenoic acid (DHA) and alpha-linoleic acid (ALA) facilitates rapid and transient phosphorylation of FFA4 expressed ectopically on the surface of HEK293 cells. However, the precise mechanisms that promote FFA4 phosphorylation remain elusive. In the current study, we examined the mechanisms behind both heterologous and homologous phosphorylation of FFA4 and set out to identify the foci of FFA4 phosphorylation. Our results demonstrate that basal and heterologous phosphorylation of FFA4 are mediated by protein kinase C (PKC), while G protein-coupled receptor kinase 6 (GRK6) plays the predominant role in DHA-mediated phosphorylation of FFA4. Furthermore, we identify Thr(347), Ser(350), and Ser(357) in the C-terminal tail as major sites of FFA4 phosphorylation. Concurrent mutation of these three sites leads to a FFA4 receptor that seemingly affects Gαq/11 signaling in a positive manner as demonstrated by heightened intracellular Ca(2+) responses following agonism with DHA. Importantly, this phosphodefective FFA4 mutant lacked the ability to promote β-arrestin-2 recruitment to the cell membrane. Since many of the functionally beneficial physiological effects of FFA4 are noted to be β-arrestin mediated, these findings could provide insight into the structural requirements for FFA4 function.
Collapse
Affiliation(s)
- Rebecca N Burns
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Monalisa Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Ilya S Senatorov
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Nader H Moniri
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA.
| |
Collapse
|
43
|
Zhang B, Albaker A, Plouffe B, Lefebvre C, Tiberi M. Constitutive activities and inverse agonism in dopamine receptors. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 70:175-214. [PMID: 24931197 DOI: 10.1016/b978-0-12-417197-8.00007-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The concept of activation in the absence of agonists has been demonstrated for many GPCRs and is now solidified as one of the principal aspects of GPCR signaling. In this chapter, we review how dopamine receptors demonstrate this ability. Although difficult to prove in vivo due to the presence of endogenous dopamine and lack of subtype-selective inverse agonists and "pure" antagonists (neutral ligands), in vitro assays such as measuring intracellular cAMP, [(35)S]GTPγS binding, and [(3)H]thymidine incorporation have uncovered the constitutive activation of D1- and D2-class receptors. Nevertheless, because of limited and inconsistent findings, the existence of constitutive activity for D2-class receptors is currently not well established. Mutagenesis studies have shown that basal signaling, notably by D1-class receptors, is governed by the collective contributions of transmembrane domains and extracellular/intracellular loops, such as the third extracellular loop, the third intracellular loop, and C-terminal tail. Furthermore, constitutive activities of D1-class receptors are subjected to regulation by kinases. Among the dopamine receptor family, the D5 receptor subtype exhibits a higher basal signaling and bears resemblance to constitutively active mutant forms of GPCRs. The presence of its constitutive activity in vivo and its pathophysiological relevance, with a brief mention of other subtypes, are also discussed.
Collapse
Affiliation(s)
- Boyang Zhang
- Ottawa Hospital Research Institute (Neuroscience Program), Ottawa, Ontario, Canada; Departments of Medicine, Cellular & Molecular Medicine, Psychiatry, University of Ottawa, Ottawa, Ontario, Canada
| | - Awatif Albaker
- Ottawa Hospital Research Institute (Neuroscience Program), Ottawa, Ontario, Canada; Departments of Medicine, Cellular & Molecular Medicine, Psychiatry, University of Ottawa, Ottawa, Ontario, Canada
| | - Bianca Plouffe
- Department of Biochemistry, Université de Montréal, Montréal, Québec, Canada; Institut de recherche en immunologie, cancer, Montréal, Québec, Canada
| | - Caroline Lefebvre
- Ottawa Hospital Research Institute (Neuroscience Program), Ottawa, Ontario, Canada; Departments of Medicine, Cellular & Molecular Medicine, Psychiatry, University of Ottawa, Ottawa, Ontario, Canada
| | - Mario Tiberi
- Ottawa Hospital Research Institute (Neuroscience Program), Ottawa, Ontario, Canada; Departments of Medicine, Cellular & Molecular Medicine, Psychiatry, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
44
|
Jeong J, Park YU, Kim DK, Lee S, Kwak Y, Lee SA, Lee H, Suh YH, Gho YS, Hwang D, Park SK. Cdk5 phosphorylates dopamine D2 receptor and attenuates downstream signaling. PLoS One 2013; 8:e84482. [PMID: 24391960 PMCID: PMC3877277 DOI: 10.1371/journal.pone.0084482] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 11/14/2013] [Indexed: 11/19/2022] Open
Abstract
The dopamine D2 receptor (DRD2) is a key receptor that mediates dopamine-associated brain functions such as mood, reward, and emotion. Cyclin-dependent kinase 5 (Cdk5) is a proline-directed serine/threonine kinase whose function has been implicated in the brain reward circuit. In this study, we revealed that the serine 321 residue (S321) in the third intracellular loop of DRD2 (D2i3) is a novel regulatory site of Cdk5. Cdk5-dependent phosphorylation of S321 in the D2i3 was observed in in vitro and cell culture systems. We further observed that the phosphorylation of S321 impaired the agonist-stimulated surface expression of DRD2 and decreased G protein coupling to DRD2. Moreover, the downstream cAMP pathway was affected in the heterologous system and in primary neuronal cultures from p35 knockout embryos likely due to the reduced inhibitory activity of DRD2. These results indicate that Cdk5-mediated phosphorylation of S321 inhibits DRD2 function, providing a novel regulatory mechanism for dopamine signaling.
Collapse
Affiliation(s)
- Jaehoon Jeong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Young-Un Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Dae-Kyum Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Saebom Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Yongdo Kwak
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Seol-Ae Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Haeryun Lee
- Division of Integrative Bioscience and Biotechnology, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Yoo-Hun Suh
- Korea Brain Research Institute, Daegu, Republic of Korea
| | - Yong Song Gho
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Daehee Hwang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Sang Ki Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
- * E-mail:
| |
Collapse
|
45
|
Basu D, Tian Y, Bhandari J, Jiang JR, Hui P, Johnson RL, Mishra RK. Effects of the dopamine D2 allosteric modulator, PAOPA, on the expression of GRK2, arrestin-3, ERK1/2, and on receptor internalization. PLoS One 2013; 8:e70736. [PMID: 23940634 PMCID: PMC3735488 DOI: 10.1371/journal.pone.0070736] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 06/26/2013] [Indexed: 11/19/2022] Open
Abstract
The activity of G protein-coupled receptors (GPCRs) is intricately regulated by a range of intracellular proteins, including G protein-coupled kinases (GRKs) and arrestins. Understanding the effects of ligands on these signaling pathways could provide insights into disease pathophysiologies and treatment. The dopamine D2 receptor is a GPCR strongly implicated in the pathophysiology of a range of neurological and neuropsychiatric disorders, particularly schizophrenia. Previous studies from our lab have shown the preclinical efficacy of a novel allosteric drug, 3(R)- [(2(S)-pyrrolidinylcarbonyl)amino]-2-oxo-1-pyrrolidineacetamide (PAOPA), in attenuating schizophrenia-like behavioural abnormalities in rodent models of the disease. As an allosteric modulator, PAOPA binds to a site on the D2 receptor, which is distinct from the endogenous ligand-binding site, in order to modulate the binding of the D2 receptor ligand, dopamine. The exact signaling pathways affected by this allosteric modulator are currently unknown. The objectives of this study were to decipher the in vivo effects, in rats, of chronic PAOPA administration on D2 receptor regulatory and downstream molecules, including GRK2, arrestin-3 and extracellular receptor kinase (ERK) 1/2. Additionally, an in vitro cellular model was also used to study PAOPA’s effects on D2 receptor internalization. Results from western immunoblots showed that chronic PAOPA treatment increased the striatal expression of GRK2 by 41%, arrestin-3 by 34%, phospho-ERK1 by 51% and phospho-ERK2 by 36%. Results also showed that the addition of PAOPA to agonist treatment in cells increased D2 receptor internalization by 33%. This study provides the foundational evidence of putative signaling pathways, and changes in receptor localization, affected by treatment with PAOPA. It improves our understanding on the diverse mechanisms of action of allosteric modulators, while advancing PAOPA’s development into a novel drug for the improved treatment of schizophrenia.
Collapse
Affiliation(s)
- Dipannita Basu
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Yuxin Tian
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Jayant Bhandari
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Jian Ru Jiang
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Patricia Hui
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Rodney L. Johnson
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Ram K. Mishra
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
- * E-mail:
| |
Collapse
|
46
|
Celver J, Sharma M, Thanawala V, Christopher Octeau J, Kovoor A. Arrestin-dependent but G-protein coupled receptor kinase-independent uncoupling of D2-dopamine receptors. J Neurochem 2013; 127:57-65. [PMID: 23815307 DOI: 10.1111/jnc.12359] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 06/26/2013] [Accepted: 06/28/2013] [Indexed: 11/30/2022]
Abstract
We reconstituted D2 like dopamine receptor (D2R) and the delta opioid receptor (DOR) coupling to G-protein gated inwardly rectifying potassium channels (K(ir)3) and directly compared the effects of co-expression of G-protein coupled receptor kinase (GRK) and arrestin on agonist-dependent desensitization of the receptor response. We found, as described previously, that co-expression of a GRK and an arrestin synergistically increased the rate of agonist-dependent desensitization of DOR. In contrast, only arrestin expression was required to produce desensitization of D2R responses. Furthermore, arrestin-dependent GRK-independent desensitization of D2R-K(ir)3 coupling could be transferred to DOR by substituting the third cytoplasmic loop of DOR with that of D2R. The arrestin-dependent GRK-independent desensitization of D2R desensitization was inhibited by staurosporine treatment, and blocked by alanine substitution of putative protein kinase C phosphorylation sites in the third cytoplasmic loop of D2R. Finally, the D2R construct in which putative protein kinase C phosphorylation sites were mutated did not undergo significant agonist-dependent desensitization even after GRK co-expression, suggesting that GRK phosphorylation of D2R does not play an important role in uncoupling of the receptor.
Collapse
Affiliation(s)
- Jeremy Celver
- Department of Biomedical and Pharmacological Sciences, College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| | | | | | | | | |
Collapse
|
47
|
Ustione A, Piston DW, Harris PE. Minireview: Dopaminergic regulation of insulin secretion from the pancreatic islet. Mol Endocrinol 2013; 27:1198-207. [PMID: 23744894 DOI: 10.1210/me.2013-1083] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Exogenous dopamine inhibits insulin secretion from pancreatic β-cells, but the lack of dopaminergic neurons in pancreatic islets has led to controversy regarding the importance of this effect. Recent data, however, suggest a plausible physiologic role for dopamine in the regulation of insulin secretion. We review the literature underlying our current understanding of dopaminergic signaling that can down-regulate glucose-stimulated insulin secretion from pancreatic islets. In this negative feedback loop, dopamine is synthesized in the β-cells from circulating L-dopa, serves as an autocrine signal that is cosecreted with insulin, and causes a tonic inhibition on glucose-stimulated insulin secretion. On the whole animal scale, L-dopa is produced by cells in the gastrointestinal tract, and its concentration in the blood plasma increases following a mixed meal. By reviewing the outcome of certain types of bariatric surgery that result in rapid amelioration of glucose tolerance, we hypothesize that dopamine serves as an "antiincretin" signal that counterbalances the stimulatory effect of glucagon-like peptide 1.
Collapse
Affiliation(s)
- Alessandro Ustione
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, Tennessee 37232-0615, USA
| | | | | |
Collapse
|
48
|
Hozumi Y, Watanabe M, Goto K. Signaling cascade of diacylglycerol kinase β in the pituitary intermediate lobe: dopamine D2 receptor/phospholipase Cβ4/diacylglycerol kinase β/protein kinase Cα. J Histochem Cytochem 2013; 58:119-29. [PMID: 19826069 DOI: 10.1369/jhc.2009.954347] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Accepted: 09/21/2009] [Indexed: 02/05/2023] Open
Abstract
The pituitary gland dynamically changes its hormone output under various pathophysiological conditions. One of the pathways implicated in the regulatory mechanism of this gland is a dopaminergic system that operates the phosphoinositide (PI) cycle to transmit downstream signal through second messengers. We have previously shown that diacylglycerol kinase β (DGKβ) is coexpressed with dopamine D1 and D2 receptors in medium spiny neurons of the striatum, suggesting a plausible implication of DGKβ in dopaminergic transmission. However, it remains elusive whether DGKβ is involved in the dopaminergic system in the pituitary gland. The aim of this study is to investigate the expression and localization of DGK in the pituitary gland, together with the molecular components involved in the PI signaling cascade, including dopamine receptors, phospholipase C (PLC), and a major downstream molecule, protein kinase C (PKC). Here we show that DGKβ and the dopamine D2 receptor are coexpressed in the intermediate lobe and localize to the plasma membrane side by side. In addition, we reveal that PLCβ4 and PKCα are the subtypes expressed in the intermediate lobe among those families. These findings will substantiate and further extend our understanding of the molecular-anatomical pathway of PI signaling and the functional roles of DGK in the pituitary intermediate lobe.
Collapse
Affiliation(s)
- Yasukazu Hozumi
- Department of Anatomy and Cell Biology, Yamagata University School of Medicine, Yamagata 990-9585, Japan.
| | | | | |
Collapse
|
49
|
Chen R, Daining CP, Sun H, Fraser R, Stokes SL, Leitges M, Gnegy ME. Protein kinase Cβ is a modulator of the dopamine D2 autoreceptor-activated trafficking of the dopamine transporter. J Neurochem 2013; 125:663-72. [PMID: 23458603 DOI: 10.1111/jnc.12229] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 02/13/2013] [Accepted: 02/25/2013] [Indexed: 11/28/2022]
Abstract
The strength and duration of extracellular dopamine concentrations are regulated by the presynaptic dopamine transporter (DAT) and dopamine D2 autoreceptors (D2autoRs). There is a functional interaction between these two proteins. Activation of D2autoRs increases DAT trafficking to the surface whereas disruption of this interaction compromises activities of both proteins and alters dopaminergic transmission. Previously we reported that DAT expression and activity are subject to modulation by protein kinase Cβ (PKCβ). Here, we further demonstrate that PKCβ is integral for the interaction between DAT and D2autoR. Inhibition or absence of PKCβ abolished the communication between DAT and D2autoR. In mouse striatal synaptosomes and transfected N2A cells, the D2autoR-stimulated membrane insertion of DAT was abolished by PKCβ inhibition. Moreover, D2autoR-stimulated DAT trafficking is mediated by a PKCβ-extracellular signal-regulated kinase signaling cascade where PKCβ is upstream of extracellular signal-regulated kinase. The increased surface DAT expression upon D2autoR activation resulted from enhanced DAT recycling as opposed to reduced internalization. Further, PKCβ promoted accelerated DAT recycling. Our study demonstrates that PKCβ critically regulates D2autoR-activated DAT trafficking and dopaminergic signaling. PKCβ is a potential drug target for correcting abnormal extracellular dopamine levels in diseases such as drug addiction and schizophrenia.
Collapse
Affiliation(s)
- Rong Chen
- Department of Physiology & Pharmacology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Cho DI, Zheng M, Min C, Kwon KJ, Shin CY, Choi HK, Kim KM. ARF6 and GASP-1 are post-endocytic sorting proteins selectively involved in the intracellular trafficking of dopamine D₂ receptors mediated by GRK and PKC in transfected cells. Br J Pharmacol 2013; 168:1355-74. [PMID: 23082996 PMCID: PMC3596642 DOI: 10.1111/bph.12025] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 09/07/2012] [Accepted: 09/28/2012] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE GPCRs undergo both homologous and heterologous regulatory processes in which receptor phosphorylation plays a critical role. The protein kinases responsible for each pathway are well established; however, other molecular details that characterize each pathway remain unclear. In this study, the molecular mechanisms that determine the differences in the functional roles and intracellular trafficking between homologous and PKC-mediated heterologous internalization pathways for the dopamine D₂ receptor were investigated. EXPERIMENTAL APPROACH All of the S/T residues located within the intracellular loops of D₂ receptor were mutated, and the residues responsible for GRK- and PKC-mediated internalization were determined in HEK-293 cells and SH-SY5Y cells. The functional role of receptor internalization and the cellular components that determine the post-endocytic fate of internalized D₂ receptors were investigated in the transfected cells. KEY RESULTS T134, T225/S228/S229 and S325 were involved in PKC-mediated D₂ receptor desensitization. S229 and adjacent S/T residues mediated the PKC-dependent internalization of D₂ receptors, which induced down-regulation and desensitization. S/T residues within the second intracellular loop and T225 were the major residues involved in GRK-mediated internalization of D₂ receptors, which induced receptor resensitization. ARF6 mediated the recycling of D₂ receptors internalized in response to agonist stimulation. In contrast, GASP-1 mediated the down-regulation of D₂ receptors internalized in a PKC-dependent manner. CONCLUSIONS AND IMPLICATIONS GRK- and PKC-mediated internalizations of D₂ receptors occur through different intracellular trafficking pathways and mediate distinct functional roles. Distinct S/T residues within D₂ receptors and different sorting proteins are involved in the dissimilar regulation of D₂ receptors by GRK2 and PKC.
Collapse
Affiliation(s)
- D I Cho
- Department of Pharmacology, College of Pharmacy, Drug Development Research Institute, Chonnam National University, Gwang-Ju, Korea
| | | | | | | | | | | | | |
Collapse
|