1
|
Luppi AI, Singleton SP, Hansen JY, Jamison KW, Bzdok D, Kuceyeski A, Betzel RF, Misic B. Contributions of network structure, chemoarchitecture and diagnostic categories to transitions between cognitive topographies. Nat Biomed Eng 2024; 8:1142-1161. [PMID: 39103509 PMCID: PMC11410673 DOI: 10.1038/s41551-024-01242-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/02/2024] [Indexed: 08/07/2024]
Abstract
The mechanisms linking the brain's network structure to cognitively relevant activation patterns remain largely unknown. Here, by leveraging principles of network control, we show how the architecture of the human connectome shapes transitions between 123 experimentally defined cognitive activation maps (cognitive topographies) from the NeuroSynth meta-analytic database. Specifically, we systematically integrated large-scale multimodal neuroimaging data from functional magnetic resonance imaging, diffusion tractography, cortical morphometry and positron emission tomography to simulate how anatomically guided transitions between cognitive states can be reshaped by neurotransmitter engagement or by changes in cortical thickness. Our model incorporates neurotransmitter-receptor density maps (18 receptors and transporters) and maps of cortical thickness pertaining to a wide range of mental health, neurodegenerative, psychiatric and neurodevelopmental diagnostic categories (17,000 patients and 22,000 controls). The results provide a comprehensive look-up table charting how brain network organization and chemoarchitecture interact to manifest different cognitive topographies, and establish a principled foundation for the systematic identification of ways to promote selective transitions between cognitive topographies.
Collapse
Affiliation(s)
- Andrea I Luppi
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| | - S Parker Singleton
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
| | - Justine Y Hansen
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Keith W Jamison
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
| | - Danilo Bzdok
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
- MILA, Quebec Artificial Intelligence Institute, Montreal, Quebec, Canada
| | - Amy Kuceyeski
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Richard F Betzel
- Psychological and Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Bratislav Misic
- Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
2
|
Maaliki D, Jaffa AA, Nasser S, Sahebkar A, Eid AH. Adrenoceptor Desensitization: Current Understanding of Mechanisms. Pharmacol Rev 2024; 76:358-387. [PMID: 38697858 DOI: 10.1124/pharmrev.123.000831] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 05/05/2024] Open
Abstract
G-protein coupled receptors (GPCRs) transduce a wide range of extracellular signals. They are key players in the majority of biologic functions including vision, olfaction, chemotaxis, and immunity. However, as essential as most of them are to body function and homeostasis, overactivation of GPCRs has been implicated in many pathologic diseases such as cancer, asthma, and heart failure (HF). Therefore, an important feature of G protein signaling systems is the ability to control GPCR responsiveness, and one key process to control overstimulation involves initiating receptor desensitization. A number of steps are appreciated in the desensitization process, including cell surface receptor phosphorylation, internalization, and downregulation. Rapid or short-term desensitization occurs within minutes and involves receptor phosphorylation via the action of intracellular protein kinases, the binding of β-arrestins, and the consequent uncoupling of GPCRs from their cognate heterotrimeric G proteins. On the other hand, long-term desensitization occurs over hours to days and involves receptor downregulation or a decrease in cell surface receptor protein level. Of the proteins involved in this biologic phenomenon, β-arrestins play a particularly significant role in both short- and long-term desensitization mechanisms. In addition, β-arrestins are involved in the phenomenon of biased agonism, where the biased ligand preferentially activates one of several downstream signaling pathways, leading to altered cellular responses. In this context, this review discusses the different patterns of desensitization of the α 1-, α 2- and the β adrenoceptors and highlights the role of β-arrestins in regulating physiologic responsiveness through desensitization and biased agonism. SIGNIFICANCE STATEMENT: A sophisticated network of proteins orchestrates the molecular regulation of GPCR activity. Adrenoceptors are GPCRs that play vast roles in many physiological processes. Without tightly controlled desensitization of these receptors, homeostatic imbalance may ensue, thus precipitating various diseases. Here, we critically appraise the mechanisms implicated in adrenoceptor desensitization. A better understanding of these mechanisms helps identify new druggable targets within the GPCR desensitization machinery and opens exciting therapeutic fronts in the treatment of several pathologies.
Collapse
Affiliation(s)
- Dina Maaliki
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon (D.M.); School of Medicine, University of South Carolina, Columbia, South Carolina (A.A.J.); Keele University, Staffordshire, United Kingdom (S.N.); Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Aneese A Jaffa
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon (D.M.); School of Medicine, University of South Carolina, Columbia, South Carolina (A.A.J.); Keele University, Staffordshire, United Kingdom (S.N.); Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Suzanne Nasser
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon (D.M.); School of Medicine, University of South Carolina, Columbia, South Carolina (A.A.J.); Keele University, Staffordshire, United Kingdom (S.N.); Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Amirhossein Sahebkar
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon (D.M.); School of Medicine, University of South Carolina, Columbia, South Carolina (A.A.J.); Keele University, Staffordshire, United Kingdom (S.N.); Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Ali H Eid
- Department of Pharmacology and Toxicology, American University of Beirut, Beirut, Lebanon (D.M.); School of Medicine, University of South Carolina, Columbia, South Carolina (A.A.J.); Keele University, Staffordshire, United Kingdom (S.N.); Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran (A.S.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| |
Collapse
|
3
|
Saggu S, Chen Y, Cottingham C, Rehman H, Wang H, Zhang S, Augelli-Szafran C, Lu S, Lambert N, Jiao K, Lu XY, Wang Q. Activation of a novel α 2AAR-spinophilin-cofilin axis determines the effect of α 2 adrenergic drugs on fear memory reconsolidation. Mol Psychiatry 2023; 28:588-600. [PMID: 36357671 PMCID: PMC9647772 DOI: 10.1038/s41380-022-01851-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 10/12/2022] [Accepted: 10/20/2022] [Indexed: 11/11/2022]
Abstract
Posttraumatic stress disorder (PTSD) after the pandemic has emerged as a major neuropsychiatric component of post-acute COVID-19 syndrome, yet the current pharmacotherapy for PTSD is limited. The use of adrenergic drugs to treat PTSD has been suggested; however, it is hindered by conflicting clinical results and a lack of mechanistic understanding of drug actions. Our studies, using both genetically modified mice and human induced pluripotent stem cell-derived neurons, reveal a novel α2A adrenergic receptor (α2AAR)-spinophilin-cofilin axis in the hippocampus that is critical for regulation of contextual fear memory reconsolidation. In addition, we have found that two α2 ligands, clonidine and guanfacine, exhibit differential abilities in activating this signaling axis to disrupt fear memory reconsolidation. Stimulation of α2AAR with clonidine, but not guanfacine, promotes the interaction of the actin binding protein cofilin with the receptor and with the dendritic spine scaffolding protein spinophilin to induce cofilin activation at the synapse. Spinophilin-dependent regulation of cofilin is required for clonidine-induced disruption of contextual fear memory reconsolidation. Our results inform the interpretation of differential clinical observations of these two drugs on PTSD and suggest that clonidine could provide immediate treatment for PTSD symptoms related to the current pandemic. Furthermore, our study indicates that modulation of dendritic spine morphology may represent an effective strategy for the development of new pharmacotherapies for PTSD.
Collapse
Affiliation(s)
- Shalini Saggu
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Yunjia Chen
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Christopher Cottingham
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Department of Biology, University of North Alabama, Florence, AL, 35632, USA
| | - Hasibur Rehman
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Hongxia Wang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Sixue Zhang
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL, 35205, USA
| | - Corinne Augelli-Szafran
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL, 35205, USA
- Scientific Platforms, Southern Research, Birmingham, AL, 35205, USA
| | - Sumin Lu
- Department of Pharmacology, Medical College of Georgia at Augusta University, Augusta, GA, GA30912, USA
| | - Nevin Lambert
- Department of Pharmacology, Medical College of Georgia at Augusta University, Augusta, GA, GA30912, USA
| | - Kai Jiao
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, GA, GA30912, USA
| | - Xin-Yun Lu
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Qin Wang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
4
|
Chappell K, Ait Tayeb AEK, Colle R, Bouligand J, El-Asmar K, Gressier F, Trabado S, David DJ, Feve B, Becquemont L, Corruble E, Verstuyft C. The association of ARRB1 polymorphisms with response to antidepressant treatment in depressed patients. Front Pharmacol 2022; 13:974570. [PMID: 36386175 PMCID: PMC9644891 DOI: 10.3389/fphar.2022.974570] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 10/11/2022] [Indexed: 09/02/2023] Open
Abstract
Introduction: β-arrestin 1, a protein encoded by ARRB1 involved in receptor signaling, is a potential biomarker for the response to antidepressant drug (ATD) treatment in depression. We examined ARRB1 genetic variants for their association with response following ATD treatment in METADAP, a cohort of 6-month ATD-treated depressed patients. Methods: Patients (n = 388) were assessed at baseline (M0) and after 1 (M1), 3 (M3), and 6 months (M6) of treatment for Hamilton Depression Rating Scale (HDRS) changes, response, and remission. Whole-gene ARRB1 variants identified from high-throughput sequencing were separated by a minor allele frequency (MAF)≥5%. Frequent variants (i.e., MAF≥5%) annotated by RegulomeDB as likely affecting transcription factor binding were analyzed using mixed-effects models. Rare variants (i.e., MAF<5%) were analyzed using a variant set analysis. Results: The variant set analysis of rare variants was significant in explaining HDRS score changes (T = 878.9; p = 0.0033) and remission (T = -1974.1; p = 0.034). Rare variant counts were significant in explaining response (p = 0.016), remission (p = 0.022), and HDRS scores at M1 (p = 0.0021) and M3 (p=<0.001). rs553664 and rs536852 were significantly associated with the HDRS score (rs553664: p = 0.0055 | rs536852: p = 0.046) and remission (rs553664: p = 0.026 | rs536852: p = 0.012) through their interactions with time. At M6, significantly higher HDRS scores were observed in rs553664 AA homozygotes (13.98 ± 1.06) compared to AG heterozygotes (10.59 ± 0.86; p = 0.014) and in rs536852 GG homozygotes (14.88 ± 1.10) compared to AG heterozygotes (11.26 ± 0.95; p = 0.0061). Significantly lower remitter rates were observed in rs536852 GG homozygotes (8%, n = 56) compared to AG heterozygotes (42%, n = 105) at M6 (p = 0.0018). Conclusion: Our results suggest ARRB1 variants may influence the response to ATD treatment in depressed patients. Further analysis of functional ARRB1 variants and rare variant burden in other populations would help corroborate our exploratory analysis. β-arrestin 1 and genetic variants of ARRB1 may be useful clinical biomarkers for clinical improvement following ATD treatment in depressed individuals. Clinical Trial Registration: clinicaltrials.gov; identifier NCT00526383.
Collapse
Affiliation(s)
- Kenneth Chappell
- Université Paris-Saclay, UMR 1018, CESP-Inserm, Team MOODS, Faculté de Pharmacie, Bâtiment Henri MOISSAN, Orsay, France
| | - Abd El Kader Ait Tayeb
- Université Paris-Saclay, UMR 1018, CESP-Inserm, Team MOODS, Faculté de Pharmacie, Bâtiment Henri MOISSAN, Orsay, France
- Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Romain Colle
- Université Paris-Saclay, UMR 1018, CESP-Inserm, Team MOODS, Faculté de Pharmacie, Bâtiment Henri MOISSAN, Orsay, France
- Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Jérôme Bouligand
- INSERM UMR-S U1185, Faculté de Médecine, University Paris-Saclay, Le Kremlin Bicêtre, France
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Khalil El-Asmar
- Université Paris-Saclay, UMR 1018, CESP-Inserm, Team MOODS, Faculté de Pharmacie, Bâtiment Henri MOISSAN, Orsay, France
- Department of Epidemiology and Population Health, Faculty of Health Sciences, American University of Beirut, Beirut, Lebanon
| | - Florence Gressier
- Université Paris-Saclay, UMR 1018, CESP-Inserm, Team MOODS, Faculté de Pharmacie, Bâtiment Henri MOISSAN, Orsay, France
- Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Séverine Trabado
- INSERM UMR-S U1185, Faculté de Médecine, University Paris-Saclay, Le Kremlin Bicêtre, France
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Denis Joseph David
- Université Paris-Saclay, UMR 1018, CESP-Inserm, Team MOODS, Faculté de Pharmacie, Bâtiment Henri MOISSAN, Orsay, France
- CESP, MOODS Team, INSERM UMR 1018, Faculté de Médecine, University Paris-Saclay, Le Kremlin Bicêtre, France
| | - Bruno Feve
- Sorbonne Université-INSERM, Centre de Recherche Saint-Antoine, UMR S938, Institut Hospitalo-Universitaire ICAN, Service d’Endocrinologie, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Centre de Référence des Maladies Rares de l’Insulino-Sécrétion et de l’Insulino-Sensibilité, Paris, France
| | - Laurent Becquemont
- Université Paris-Saclay, UMR 1018, CESP-Inserm, Team MOODS, Faculté de Pharmacie, Bâtiment Henri MOISSAN, Orsay, France
- Centre de Recherche Clinique Paris-Saclay, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| | - Emmanuelle Corruble
- Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
- CESP, MOODS Team, INSERM UMR 1018, Faculté de Médecine, University Paris-Saclay, Le Kremlin Bicêtre, France
| | - Céline Verstuyft
- Université Paris-Saclay, UMR 1018, CESP-Inserm, Team MOODS, Faculté de Pharmacie, Bâtiment Henri MOISSAN, Orsay, France
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
- Centre de Ressources Biologiques Paris-Saclay, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Hôpital de Bicêtre, Le Kremlin Bicêtre, France
| |
Collapse
|
5
|
Aslanoglou D, Bertera S, Friggeri L, Sánchez-Soto M, Lee J, Xue X, Logan RW, Lane JR, Yechoor VK, McCormick PJ, Meiler J, Free RB, Sibley DR, Bottino R, Freyberg Z. Dual pancreatic adrenergic and dopaminergic signaling as a therapeutic target of bromocriptine. iScience 2022; 25:104771. [PMID: 35982797 PMCID: PMC9379584 DOI: 10.1016/j.isci.2022.104771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 06/10/2022] [Accepted: 07/11/2022] [Indexed: 11/23/2022] Open
Abstract
Bromocriptine is approved as a diabetes therapy, yet its therapeutic mechanisms remain unclear. Though bromocriptine's actions have been mainly attributed to the stimulation of brain dopamine D2 receptors (D2R), bromocriptine also targets the pancreas. Here, we employ bromocriptine as a tool to elucidate the roles of catecholamine signaling in regulating pancreatic hormone secretion. In β-cells, bromocriptine acts on D2R and α2A-adrenergic receptor (α2A-AR) to reduce glucose-stimulated insulin secretion (GSIS). Moreover, in α-cells, bromocriptine acts via D2R to reduce glucagon secretion. α2A-AR activation by bromocriptine recruits an ensemble of G proteins with no β-arrestin2 recruitment. In contrast, D2R recruits G proteins and β-arrestin2 upon bromocriptine stimulation, demonstrating receptor-specific signaling. Docking studies reveal distinct bromocriptine binding to α2A-AR versus D2R, providing a structural basis for bromocriptine's dual actions on β-cell α2A-AR and D2R. Together, joint dopaminergic and adrenergic receptor actions on α-cell and β-cell hormone release provide a new therapeutic mechanism to improve dysglycemia.
Collapse
Affiliation(s)
- Despoina Aslanoglou
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Suzanne Bertera
- Institute of Cellular Therapeutics, Allegheny Health Network Research Institute, Allegheny Health Network, Pittsburgh, PA, USA
| | - Laura Friggeri
- Department of Chemistry, Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
| | - Marta Sánchez-Soto
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jeongkyung Lee
- Diabetes and Beta Cell Biology Center, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiangning Xue
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ryan W. Logan
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, MA, USA
| | - 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 Protein and Receptors, Universities of Birmingham and Nottingham, Nottingham, UK
| | - Vijay K. Yechoor
- Diabetes and Beta Cell Biology Center, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peter J. McCormick
- Centre for Endocrinology, William Harvey Research Institute, Bart’s and the London School of Medicine and Dentistry, Queen Mary, University of London, London, UK
| | - Jens Meiler
- Department of Chemistry, Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
- Institute for Drug Discovery, Leipzig University Medical School, Leipzig, Germany
| | - R. Benjamin Free
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - David R. Sibley
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Rita Bottino
- Institute of Cellular Therapeutics, Allegheny Health Network Research Institute, Allegheny Health Network, Pittsburgh, PA, USA
- Imagine Pharma, Pittsburgh, PA, USA
| | - Zachary Freyberg
- Translational Neuroscience Program, Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Cell Biology, University of Pittsburgh, PA, USA
| |
Collapse
|
6
|
Saggu S, Chen Y, Chen L, Pizarro D, Pati S, Law WJ, McMahon L, Jiao K, Wang Q. A peptide blocking the ADORA1-neurabin interaction is anticonvulsant and inhibits epilepsy in an Alzheimer's model. JCI Insight 2022; 7:155002. [PMID: 35674133 PMCID: PMC9220929 DOI: 10.1172/jci.insight.155002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 04/20/2022] [Indexed: 11/17/2022] Open
Abstract
Epileptic seizures are common sequelae of stroke, acute brain injury, and chronic neurodegenerative diseases, including Alzheimer's disease (AD), and cannot be effectively controlled in approximately 40% of patients, necessitating the development of novel therapeutic agents. Activation of the A1 receptor (A1R) by endogenous adenosine is an intrinsic mechanism to self-terminate seizures and protect neurons from excitotoxicity. However, targeting A1R for neurological disorders has been hindered by side effects associated with its broad expression outside the nervous system. Here we aim to target the neural-specific A1R/neurabin/regulator of G protein signaling 4 (A1R/neurabin/RGS4) complex that dictates A1R signaling strength and response outcome in the brain. We developed a peptide that blocks the A1R-neurabin interaction to enhance A1R activity. Intracerebroventricular or i.n. administration of this peptide shows marked protection against kainate-induced seizures and neuronal death. Furthermore, in an AD mouse model with spontaneous seizures, nasal delivery of this blocking peptide reduces epileptic spike frequency. Significantly, the anticonvulsant and neuroprotective effects of this peptide are achieved through enhanced A1R function in response to endogenous adenosine in the brain, thus, avoiding side effects associated with A1R activation in peripheral tissues and organs. Our study informs potentially new anti-seizure therapy applicable to epilepsy and other neurological illness with comorbid seizures.
Collapse
Affiliation(s)
- Shalini Saggu
- Departments of Cell, Developmental and Integrative Biology
| | - Yunjia Chen
- Departments of Cell, Developmental and Integrative Biology
| | - Liping Chen
- Departments of Cell, Developmental and Integrative Biology
| | | | | | - Wen Jing Law
- Departments of Cell, Developmental and Integrative Biology
| | - Lori McMahon
- Departments of Cell, Developmental and Integrative Biology
| | - Kai Jiao
- Department of Genetics, University of Alabama at Birmingham, Alabama, USA
| | - Qin Wang
- Departments of Cell, Developmental and Integrative Biology
| |
Collapse
|
7
|
Sandrini L, Amadio P, Ieraci A, Malara A, Werba JP, Soprano PM, Balduini A, Zarà M, Bonomi A, Veglia F, Colombo GI, Popoli M, Lee FS, Tremoli E, Barbieri SS. The α 2-adrenergic receptor pathway modulating depression influences the risk of arterial thrombosis associated with BDNFVal66Met polymorphism. Biomed Pharmacother 2021; 146:112557. [PMID: 34965503 DOI: 10.1016/j.biopha.2021.112557] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/09/2021] [Accepted: 12/19/2021] [Indexed: 12/12/2022] Open
Abstract
Depression is associated with thrombotic risk and arterial events, its proper management is strongly recommended in coronary artery disease (CAD) patients. We have previously shown that the Brain-Derived Neurotrophic Factor (BDNF)Val66Met polymorphism, related to depression, is associated with arterial thrombosis in mice, and with an increased risk of acute myocardial infarction in humans. Herein, expanding the previous findings on BDNFVal66Met polymorphism, we show that desipramine, a norepinephrine reuptake-inhibitor, rescues behavioral impairments, reduces the arterial thrombosis risk, abolishes pathological coagulation and platelet hyper-reactivity, normalizes leukocyte, platelet, and bone marrow megakaryocyte number and restores physiological norepinephrine levels in homozygous knock-in BDNF Val66Met (BDNFMet/Met) mice. The in vitro data confirm the enhanced procoagulant activity and the alpha2A-adrenergic receptor (α2A-ADR) overexpression found in BDNFMet/Met mice and we provide evidence that, in presence of Met variant, norepinephrine is crucial to up-regulate procoagulant activity and to enhance platelet generation. The α2-ADR antagonist rauwolscine rescues the prothrombotic phenotype in BDNFMet/Met mice and reduces procoagulant activity and platelet generation in cells transfected with BDNFMet plasmid or exposed to pro-BDNFMet peptide. Finally, we show that homozygous BDNFMet/Met CAD patients have hyper-reactive platelets overexpressing abundant α2A-ADR. The great proplatelet release from their megakaryocytes well reflects their higher circulating platelet number compared to BDNFVal/Val patients. These data reveal an unprecedented described role of Met allele in the dysregulation of norepinephrine/α2A-ADR pathway that may explain the predisposition to arterial thrombosis. Overall, the development of α2A-ADR inhibitors might represent a pharmacological treatment for depression-associated thrombotic conditions in this specific subgroup of CAD patients.
Collapse
Affiliation(s)
| | | | - Alessandro Ieraci
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Alessandro Malara
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; IRCCS San Matteo Foundation, Pavia, Italy
| | - José P Werba
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Paolo M Soprano
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; IRCCS San Matteo Foundation, Pavia, Italy
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; IRCCS San Matteo Foundation, Pavia, Italy
| | - Marta Zarà
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Alice Bonomi
- Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | | | | | - Maurizio Popoli
- Department of Pharmaceutical Sciences, University of Milan, Milan, Italy
| | - Francis S Lee
- Department of Psychiatry, Weill Cornell Medical College, New York, USA
| | - Elena Tremoli
- Centro Cardiologico Monzino, IRCCS, Milan, Italy; Maria Cecilia Hospital, Cotignola, Italy
| | | |
Collapse
|
8
|
Delcourte S, Etievant A, Haddjeri N. Role of central serotonin and noradrenaline interactions in the antidepressants' action: Electrophysiological and neurochemical evidence. PROGRESS IN BRAIN RESEARCH 2021; 259:7-81. [PMID: 33541681 DOI: 10.1016/bs.pbr.2021.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The development of antidepressant drugs, in the last 6 decades, has been associated with theories based on a deficiency of serotonin (5-HT) and/or noradrenaline (NA) systems. Although the pathophysiology of major depression (MD) is not fully understood, numerous investigations have suggested that treatments with various classes of antidepressant drugs may lead to an enhanced 5-HT and/or adapted NA neurotransmissions. In this review, particular morpho-physiological aspects of these systems are first considered. Second, principal features of central 5-HT/NA interactions are examined. In this regard, the effects of the acute and sustained antidepressant administrations on these systems are discussed. Finally, future directions including novel therapeutic strategies are proposed.
Collapse
Affiliation(s)
- Sarah Delcourte
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France
| | - Adeline Etievant
- Integrative and Clinical Neurosciences EA481, University of Bourgogne Franche-Comté, Besançon, France
| | - Nasser Haddjeri
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France.
| |
Collapse
|
9
|
Cottingham CM, Patrick T, Richards MA, Blackburn KD. Tricyclic antipsychotics promote adipogenic gene expression to potentiate preadipocyte differentiation in vitro. Hum Cell 2020; 33:502-511. [PMID: 32447572 PMCID: PMC10805149 DOI: 10.1007/s13577-020-00372-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/02/2020] [Indexed: 11/30/2022]
Abstract
Antipsychotic-induced weight gain is a well-established but poorly understood clinical phenomenon. New mechanistic insights into how antipsychotics modulate adipose physiology are sorely needed, in hopes of either devising a therapeutic intervention to ameliorate weight gain or contributing to improved design of future agents. In this study, we have hypothesized that the weight gain-associated tricyclic antipsychotics clozapine and chlorpromazine directly impact adipose tissue by potentiating adipogenic differentiation of preadipocytes. Utilizing a well-established in vitro model system (3T3-L1 preadipocyte cell line), we demonstrate that, when applied specifically during induction of adipogenic differentiation, both clozapine and chlorpromazine significantly potentiate in vitro adipogenesis, observed as morphological changes and increased intracellular lipid accumulation. These persistent effects, observed at endpoints well after the end of antipsychotic exposure, are accompanied by increased transcript- and protein-level expression of the mature adipocyte marker perilipin-1, as indicated by RT-qPCR and Western blotting, but not by further upregulation of pro-adipogenic transcription factors versus positive controls. Our findings point to a possible physiological mechanism of antipsychotic-induced hyperplasia, with potentiated expression of mature adipocyte markers enhancing the differentiation and maturation of preadipocytes.
Collapse
Affiliation(s)
- Christopher M Cottingham
- Department of Biology, University of North Alabama, UNA, One Harrison Plaza, Box 5048, Florence, AL, 35632, USA.
| | - Taylor Patrick
- Department of Biology and Chemistry, Morehead State University, 103 Lappin Hall, Morehead, KY, 40351, USA
- University of Kentucky College of Medicine, UK Medical Center MN 150, Lexington, KY, 40536, USA
| | - Morgan A Richards
- Department of Biology and Chemistry, Morehead State University, 103 Lappin Hall, Morehead, KY, 40351, USA
- School of Rehabilitation and Health Sciences, Ohio State University, 453 West 10th Avenue, Columbus, OH, 43210, USA
| | - Kirkland D Blackburn
- Department of Biology and Chemistry, Morehead State University, 103 Lappin Hall, Morehead, KY, 40351, USA
- Kentucky College of Osteopathic Medicine, 147 Sycamore Street, Pikeville, KY, 41501, USA
| |
Collapse
|
10
|
Zhang F, Gannon M, Chen Y, Yan S, Zhang S, Feng W, Tao J, Sha B, Liu Z, Saito T, Saido T, Keene CD, Jiao K, Roberson ED, Xu H, Wang Q. β-amyloid redirects norepinephrine signaling to activate the pathogenic GSK3β/tau cascade. Sci Transl Med 2020; 12:eaay6931. [PMID: 31941827 PMCID: PMC7891768 DOI: 10.1126/scitranslmed.aay6931] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/14/2019] [Indexed: 12/11/2022]
Abstract
The brain noradrenergic system is critical for normal cognition and is affected at early stages in Alzheimer's disease (AD). Here, we reveal a previously unappreciated direct role of norepinephrine signaling in connecting β-amyloid (Aβ) and tau, two key pathological components of AD pathogenesis. Our results show that Aβ oligomers bind to an allosteric site on α2A adrenergic receptor (α2AAR) to redirect norepinephrine-elicited signaling to glycogen synthase kinase 3β (GSK3β) activation and tau hyperphosphorylation. This norepinephrine-dependent mechanism sensitizes pathological GSK3β/tau activation in response to nanomolar accumulations of extracellular Aβ, which is 50- to 100-fold lower than the amount required to activate GSK3β by Aβ alone. The significance of our findings is supported by in vivo evidence in two mouse models, human tissue sample analysis, and longitudinal clinical data. Our study provides translational insights into mechanisms underlying Aβ proteotoxicity, which might have strong implications for the interpretation of Aβ clearance trial results and future drug design and for understanding the selective vulnerability of noradrenergic neurons in AD.
Collapse
Affiliation(s)
- Fang Zhang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Mary Gannon
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yunjia Chen
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Shun Yan
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sixue Zhang
- Department of Chemistry, Southern Research Institute, Birmingham, AL 35205, USA
| | - Wendy Feng
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jiahui Tao
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Bingdong Sha
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Zhenghui Liu
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Takaomi Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - C Dirk Keene
- Department of Pathology, University of Washington, Seattle, WA 98104, USA
| | - Kai Jiao
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Erik D Roberson
- Alzheimer's Disease Center, Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Huaxi Xu
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Qin Wang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
11
|
Abstract
GPCRs (G-protein [guanine nucleotide-binding protein]-coupled receptors) play a central physiological role in the regulation of cardiac function in both health and disease and thus represent one of the largest class of surface receptors targeted by drugs. Several antagonists of GPCRs, such as βARs (β-adrenergic receptors) and Ang II (angiotensin II) receptors, are now considered standard of therapy for a wide range of cardiovascular disease, such as hypertension, coronary artery disease, and heart failure. Although the mechanism of action for GPCRs was thought to be largely worked out in the 80s and 90s, recent discoveries have brought to the fore new and previously unappreciated mechanisms for GPCR activation and subsequent downstream signaling. In this review, we focus on GPCRs most relevant to the cardiovascular system and discuss traditional components of GPCR signaling and highlight evolving concepts in the field, such as ligand bias, β-arrestin-mediated signaling, and conformational heterogeneity.
Collapse
Affiliation(s)
- Jialu Wang
- From the Department of Medicine (J.W., C.G., H.A.R.)
| | | | - Howard A Rockman
- From the Department of Medicine (J.W., C.G., H.A.R.).,Department of Cell Biology (H.A.R.).,Department of Molecular Genetics and Microbiology (H.A.R.), Duke University Medical Center, Durham, NC
| |
Collapse
|
12
|
Abstract
β-arrestin1 (or arrestin2) and β-arrestin2 (or arrestin3) are ubiquitously expressed cytosolic adaptor proteins that were originally discovered for their inhibitory role in G protein-coupled receptor (GPCR) signaling through heterotrimeric G proteins. However, further biochemical characterization revealed that β-arrestins do not just "block" the activated GPCRs, but trigger endocytosis and kinase activation leading to specific signaling pathways that can be localized on endosomes. The signaling pathways initiated by β-arrestins were also found to be independent of G protein activation by GPCRs. The discovery of ligands that blocked G protein activation but promoted β-arrestin binding, or vice-versa, suggested the exciting possibility of selectively activating intracellular signaling pathways. In addition, it is becoming increasingly evident that β-arrestin-dependent signaling is extremely diverse and provokes distinct cellular responses through different GPCRs even when the same effector kinase is involved. In this review, we summarize various signaling pathways mediated by β-arrestins and highlight the physiologic effects of β-arrestin-dependent signaling.
Collapse
|
13
|
Blockade of α2-adrenergic receptors in prelimbic cortex: impact on cocaine self-administration in adult spontaneously hypertensive rats following adolescent atomoxetine treatment. Psychopharmacology (Berl) 2017; 234:2897-2909. [PMID: 28730282 PMCID: PMC5693724 DOI: 10.1007/s00213-017-4681-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 06/25/2017] [Indexed: 12/17/2022]
Abstract
RATIONALE Research with the spontaneously hypertensive rat (SHR) model of attention deficit/hyperactivity disorder demonstrated that chronic methylphenidate treatment during adolescence increased cocaine self-administration established during adulthood under a progressive ratio (PR) schedule. Compared to vehicle, chronic atomoxetine treatment during adolescence failed to increase cocaine self-administration under a PR schedule in adult SHR. OBJECTIVES We determined if enhanced noradrenergic transmission at α2-adrenergic receptors within prefrontal cortex contributes to this neutral effect of adolescent atomoxetine treatment in adult SHR. METHODS Following treatment from postnatal days 28-55 with atomoxetine (0.3 mg/kg) or vehicle, adult male SHR and control rats from Wistar-Kyoto (WKY) and Wistar (WIS) strains were trained to self-administer 0.3 mg/kg cocaine. Self-administration performance was evaluated under a PR schedule of cocaine delivery following infusion of the α2-adrenergic receptor antagonist idazoxan (0 and 10-56 μg/side) directly into prelimbic cortex. RESULTS Adult SHR attained higher PR break points and had greater numbers of active lever responses and infusions than WKY and WIS. Idazoxan dose-dependently increased PR break points and active lever responses in SHR following adolescent atomoxetine vs. vehicle treatment. Behavioral changes were negligible after idazoxan pretreatment in SHR following adolescent vehicle or in WKY and WIS following adolescent atomoxetine or vehicle. CONCLUSIONS α2-Adrenergic receptor blockade in prelimbic cortex of SHR masked the expected neutral effect of adolescent atomoxetine on adult cocaine self-administration behavior. Moreover, greater efficacy of acute idazoxan challenge in adult SHR after adolescent atomoxetine relative to vehicle is consistent with the idea that chronic atomoxetine may downregulate presynaptic α2A-adrenergic autoreceptors in SHR.
Collapse
|
14
|
Mo W, Michel MC, Lee XW, Kaumann AJ, Molenaar P. The β 3 -adrenoceptor agonist mirabegron increases human atrial force through β 1 -adrenoceptors: an indirect mechanism? Br J Pharmacol 2017; 174:2706-2715. [PMID: 28574581 DOI: 10.1111/bph.13897] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 05/21/2017] [Accepted: 05/30/2017] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND AND PURPOSE Mirabegron has been classified as a β3 -adrenoceptor agonist approved for overactive bladder syndrome. We investigated possible cardiac effects of mirabegron in the absence or presence of β-adrenoceptor subtype antagonists. In view of its phenylethanolamine structure, we investigated whether mirabegron has indirect sympathomimetic activity by using neuronal uptake blockers. EXPERIMENTAL APPROACH Right atrial trabeculae, from non-failing hearts, were paced and contractile force measured at 37°C. Single concentrations of mirabegron were added in the absence or presence of the phosphodiesterase inhibitor 3-isobutyl-1-methylxanthine (IBMX), β3 (L-748,337), β1 (CGP 20712A), β2 (ICI 118,551) -adrenoceptor antagonists, neuronal uptake inhibitors desipramine or phenoxybenzamine. KEY RESULTS Mirabegron significantly increased contractile force in human right atrium (1 μM, 7.6 ± 2.6%, n = 7; 10 μM, 10.2 ± 1.5%, n = 22 compared with (-)-isoprenaline P < 0.05). In the presence of IBMX, mirabegron (10 μM) caused a greater contraction. L-748,337 (100 nM) had no effect on the increase in contractile force caused by mirabegron (10 μM). In contrast, mirabegron (10 μM) reduced contractile force in the presence of CGP 20712A, which was not affected by L-748,337 (100 nM) or ICI 118,551 (50 nM). Mirabegron (10 μM) also reduced contractile force in the presence of desipramine or phenoxybenzamine. CONCLUSIONS AND IMPLICATIONS Mirabegron increases human atrial force through β1 - but not β3 -adrenoceptors. Desipramine and phenoxybenzamine block neuronal uptake and conceivably prevent mirabegron from releasing noradrenaline. A non-specific cardiodepressant effect is not mediated through β3 (or β2 )-adrenoceptors, consistent with lack of β3 -adrenoceptor function on human atrial contractility.
Collapse
Affiliation(s)
- Weilan Mo
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Qld, Australia.,Northside Clinical School of Medicine, The University of Queensland, The Prince Charles Hospital, Brisbane, Qld, Australia.,Critical Care Research Group, The Prince Charles Hospital, Brisbane, Qld, Australia
| | - Martin C Michel
- Department of Pharmacology, Johannes Gutenberg University, Mainz, Germany
| | - Xiang Wen Lee
- Northside Clinical School of Medicine, The University of Queensland, The Prince Charles Hospital, Brisbane, Qld, Australia
| | - Alberto J Kaumann
- Departamento de Farmacología, Facultad de Medicina, Universidad de Murcia, Campus de Espinardo, Murcia, Spain
| | - Peter Molenaar
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, Qld, Australia.,Northside Clinical School of Medicine, The University of Queensland, The Prince Charles Hospital, Brisbane, Qld, Australia.,Critical Care Research Group, The Prince Charles Hospital, Brisbane, Qld, Australia
| |
Collapse
|
15
|
Zhang F, Gannon M, Chen Y, Zhou L, Jiao K, Wang Q. The amyloid precursor protein modulates α 2A-adrenergic receptor endocytosis and signaling through disrupting arrestin 3 recruitment. FASEB J 2017. [PMID: 28646018 DOI: 10.1096/fj.201700346r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The amyloid precursor protein (APP) has long been appreciated for its role in Alzheimer's disease (AD) pathology. However, less is known about the physiologic function of APP outside of AD. Particularly, whether and how APP may regulate functions of cell surface receptors, including GPCRs, remains largely unclear. In this study, we identified a novel direct interaction between APP and the α2A-adrenergic receptor (α2AAR) that occurs at the intracellular domains of both proteins. The APP interaction with α2AAR is promoted by agonist stimulation and competes with arrestin 3 binding to the receptor. Consequently, the presence of APP attenuates α2AAR internalization and desensitization, which are arrestin-dependent processes. Furthermore, in neuroblastoma neuro-2A cells and primary superior cervical ganglion neurons, where APP is highly expressed, the lack of APP leads to a dramatic increase in plasma membrane recruitment of endogenous arrestin 3 following α2AAR activation. Concomitantly, agonist-induced internalization of α2AAR is significantly enhanced in these neuronal cells. Our study provided the first evidence that APP fine tunes GPCR signaling and trafficking. Given the important role of α2AAR in controlling norepinephrine release and response, this novel regulation of α2AAR by APP may have an impact on modulation of noradrenergic activity and sympathetic tone.-Zhang, F., Gannon, M., Chen, Y., Zhou, L., Jiao, K., Wang, Q. The amyloid precursor protein modulates α2A-adrenergic receptor endocytosis and signaling through disrupting arrestin 3 recruitment.
Collapse
Affiliation(s)
- Fang Zhang
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mary Gannon
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yunjia Chen
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lufang Zhou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kai Jiao
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Qin Wang
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA;
| |
Collapse
|
16
|
Probing the Molecular Mechanism of Human Soluble Guanylate Cyclase Activation by NO in vitro and in vivo. Sci Rep 2017; 7:43112. [PMID: 28230071 PMCID: PMC5322342 DOI: 10.1038/srep43112] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 01/19/2017] [Indexed: 11/12/2022] Open
Abstract
Soluble guanylate cyclase (sGC) is a heme-containing metalloprotein in NO-sGC-cGMP signaling. NO binds to the heme of sGC to catalyze the synthesis of the second messenger cGMP, which plays a critical role in several physiological processes. However, the molecular mechanism for sGC to mediate the NO signaling remains unclear. Here fluorophore FlAsH-EDT2 and fluorescent proteins were employed to study the NO-induced sGC activation. FlAsH-EDT2 labeling study revealed that NO binding to the H-NOX domain of sGC increased the distance between H-NOX and PAS domain and the separation between H-NOX and coiled-coil domain. The heme pocket conformation changed from “closed” to “open” upon NO binding. In addition, the NO-induced conformational change of sGC was firstly investigated in vivo through fluorescence lifetime imaging microscopy. The results both in vitro and in vivo indicated the conformational change of the catalytic domain of sGC from “open” to “closed” upon NO binding. NO binding to the heme of H-NOX domain caused breaking of Fe-N coordination bond, initiated the domain moving and conformational change, induced the allosteric effect of sGC to trigger the NO-signaling from H-NOX via PAS & coiled-coil to the catalytic domain, and ultimately stimulates the cyclase activity of sGC.
Collapse
|
17
|
Cottingham C, Che P, Zhang W, Wang H, Wang RX, Percival S, Birky T, Zhou L, Jiao K, Wang Q. Diverse arrestin-recruiting and endocytic profiles of tricyclic antipsychotics acting as direct α 2A adrenergic receptor ligands. Neuropharmacology 2016; 116:38-49. [PMID: 27956055 DOI: 10.1016/j.neuropharm.2016.12.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 11/28/2016] [Accepted: 12/08/2016] [Indexed: 01/01/2023]
Abstract
The therapeutic mechanism of action underlying many psychopharmacological agents remains poorly understood, due largely to the extreme molecular promiscuity exhibited by these agents with respect to potential central nervous system targets. Agents of the tricyclic chemical class, including both antidepressants and antipsychotics, exhibit a particularly high degree of molecular promiscuity; therefore, any clarification of how these agents interact with specific central nervous system targets is of great potential significance to the field. Here, we present evidence demonstrating that tricyclic antipsychotics appear to segregate into three distinct groups based upon their molecular interactions with the centrally-important α2A adrenergic receptor (AR). Specifically, while the α2AAR binds all antipsychotics tested with similar affinities, and none of the agents are able to induce classical heterotrimeric G protein-mediated α2AAR signaling, significant differences are observed with respect to arrestin3 recruitment and receptor endocytosis. All antipsychotics tested induce arrestin3 recruitment to the α2AAR, but with differing strengths. Both chlorpromazine and clozapine drive significant α2AAR endocytosis, but via differing clathrin-dependent and lipid raft-dependent pathways, while fluphenazine does not drive a robust response. Intriguingly, in silico molecular modeling suggests that each of the three exhibits unique characteristics in interacting with the α2AAR ligand-binding pocket. In addition to establishing these three antipsychotics as novel arrestin-biased ligands at the α2AAR, our findings provide key insights into the molecular actions of these clinically-important agents.
Collapse
Affiliation(s)
- Christopher Cottingham
- Department of Cell, Molecular and Developmental Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Department of Biology and Chemistry, Morehead State University, Morehead, KY 40351, USA
| | - Pulin Che
- Department of Cell, Molecular and Developmental Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Wei Zhang
- Southern Research Institute, Birmingham, AL 35205, USA
| | - Hongxia Wang
- Department of Cell, Molecular and Developmental Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Raymond X Wang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Stefanie Percival
- Department of Cell, Molecular and Developmental Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Tana Birky
- Department of Cell, Molecular and Developmental Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lufang Zhou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kai Jiao
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Qin Wang
- Department of Cell, Molecular and Developmental Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
18
|
Jiao K, Zeng G, Niu LN, Yang HX, Ren GT, Xu XY, Li FF, Tay FR, Wang MQ. Activation of α2A-adrenergic signal transduction in chondrocytes promotes degenerative remodelling of temporomandibular joint. Sci Rep 2016; 6:30085. [PMID: 27452863 PMCID: PMC4958971 DOI: 10.1038/srep30085] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 06/28/2016] [Indexed: 12/29/2022] Open
Abstract
This study tested whether activation of adrenoreceptors in chondrocytes has roles in degenerative remodelling of temporomandibular joint (TMJ) and to determine associated mechanisms. Unilateral anterior crossbite (UAC) was established to induce TMJ degeneration in rats. Saline vehicle, α2- and β-adrenoreceptor antagonists or agonists were injected locally into the TMJ area of UAC rats. Cartilage degeneration, subchondral bone microarchitecture and the expression of adrenoreceptors, aggrecans, matrix metalloproteinases (MMPs) and RANKL by chondrocytes were evaluated. Chondrocytes were stimulated by norepinephrine to investigate signal transduction of adrenoreceptors. Increased α2A-adrenoreceptor expression was observed in condylar cartilage of UAC rats, together with cartilage degeneration and subchondral bone loss. Norepinephrine depresses aggrecans expression but stimulates MMP-3, MMP-13 and RANKL production by chondrocytes through ERK1/2 and PKA pathway; these effects were abolished by an α2A-adrenoreceptor antagonist. Furthermore, inhibition of α2A-adrenoreceptor attenuated degenerative remodelling in the condylar cartilage and subchondral bone, as revealed by increased cartilage thickness, proteoglycans and aggrecan expression, and decreased MMP-3, MMP-13 and RANKL expressions in cartilage, increased BMD, BV/TV, and decreased Tb.Sp in subchondral bone. Conversely, activation of α2A-adrenoreceptor intensified aforementioned degenerative changes in UAC rats. It is concluded that activation of α2A-adrenergic signal in chondrocytes promotes TMJ degenerative remodelling by chondrocyte-mediated pro-catabolic activities.
Collapse
Affiliation(s)
- Kai Jiao
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, 145 Changle Western Road, Xi'an, 710032, China
| | - Guang Zeng
- Department of Dentistry, Tangdu Hospital, Forth Military Medical University, Shannxi, Xi'an, 710038, China
| | - Li-Na Niu
- State Key Laboratory of Military Stomatology, Department of Prosthodontics, School of Stomatology, Fourth Military Medical University, Changle Western Road No.145, Xi'an, 710032, China
| | - Hong-Xu Yang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, 145 Changle Western Road, Xi'an, 710032, China
| | - Gao-Tong Ren
- Undergraduate Department of Oral Science, Fourth Military Medical University, Changle Western Road No.145, Xi'an, 710032, China
| | - Xin-Yue Xu
- Undergraduate Department of Oral Science, Fourth Military Medical University, Changle Western Road No.145, Xi'an, 710032, China
| | - Fei-Fei Li
- State Key Laboratory of Military Stomatology, Department of Orthodontics, School of Stomatology, Fourth Military Medical University, 145 Changle Western Road, Xi'an, 710032, China
| | - Franklin R Tay
- The Dental College of Georgia, Augusta University, Augusta, GA, USA
| | - Mei-Qing Wang
- State Key Laboratory of Military Stomatology, Department of Oral Anatomy and Physiology and TMD, School of Stomatology, Fourth Military Medical University, 145 Changle Western Road, Xi'an, 710032, China
| |
Collapse
|
19
|
Grant MP, Cavanaugh A, Breitwieser GE. 14-3-3 Proteins Buffer Intracellular Calcium Sensing Receptors to Constrain Signaling. PLoS One 2015; 10:e0136702. [PMID: 26317416 PMCID: PMC4552738 DOI: 10.1371/journal.pone.0136702] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 08/06/2015] [Indexed: 12/04/2022] Open
Abstract
Calcium sensing receptors (CaSR) interact with 14-3-3 binding proteins at a carboxyl terminal arginine-rich motif. Mutations identified in patients with familial hypocalciuric hypercalcemia, autosomal dominant hypocalcemia, pancreatitis or idiopathic epilepsy support the functional importance of this motif. We combined total internal reflection fluorescence microscopy and biochemical approaches to determine the mechanism of 14-3-3 protein regulation of CaSR signaling. Loss of 14-3-3 binding caused increased basal CaSR signaling and plasma membrane levels, and a significantly larger signaling-evoked increase in plasma membrane receptors. Block of core glycosylation with tunicamycin demonstrated that changes in plasma membrane CaSR levels were due to differences in exocytic rate. Western blotting to quantify time-dependent changes in maturation of expressed wt CaSR and a 14-3-3 protein binding-defective mutant demonstrated that signaling increases synthesis to maintain constant levels of the immaturely and maturely glycosylated forms. CaSR thus operates by a feed-forward mechanism, whereby signaling not only induces anterograde trafficking of nascent receptors but also increases biosynthesis to maintain steady state levels of net cellular CaSR. Overall, these studies suggest that 14-3-3 binding at the carboxyl terminus provides an important buffering mechanism to increase the intracellular pool of CaSR available for signaling-evoked trafficking, but attenuates trafficking to control the dynamic range of responses to extracellular calcium.
Collapse
Affiliation(s)
- Michael P. Grant
- Weis Center for Research, Geisinger Clinic, Danville, Pennsylvania, United States of America
| | - Alice Cavanaugh
- Weis Center for Research, Geisinger Clinic, Danville, Pennsylvania, United States of America
| | - Gerda E. Breitwieser
- Weis Center for Research, Geisinger Clinic, Danville, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
20
|
RGS9-2--controlled adaptations in the striatum determine the onset of action and efficacy of antidepressants in neuropathic pain states. Proc Natl Acad Sci U S A 2015; 112:E5088-97. [PMID: 26305935 DOI: 10.1073/pnas.1504283112] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The striatal protein Regulator of G-protein signaling 9-2 (RGS9-2) plays a key modulatory role in opioid, monoamine, and other G-protein-coupled receptor responses. Here, we use the murine spared-nerve injury model of neuropathic pain to investigate the mechanism by which RGS9-2 in the nucleus accumbens (NAc), a brain region involved in mood, reward, and motivation, modulates the actions of tricyclic antidepressants (TCAs). Prevention of RGS9-2 action in the NAc increases the efficacy of the TCA desipramine and dramatically accelerates its onset of action. By controlling the activation of effector molecules by G protein α and βγ subunits, RGS9-2 affects several protein interactions, phosphoprotein levels, and the function of the epigenetic modifier histone deacetylase 5, which are important for TCA responsiveness. Furthermore, information from RNA-sequencing analysis reveals that RGS9-2 in the NAc affects the expression of many genes known to be involved in nociception, analgesia, and antidepressant drug actions. Our findings provide novel information on NAc-specific cellular mechanisms that mediate the actions of TCAs in neuropathic pain states.
Collapse
|
21
|
Gannon M, Che P, Chen Y, Jiao K, Roberson ED, Wang Q. Noradrenergic dysfunction in Alzheimer's disease. Front Neurosci 2015; 9:220. [PMID: 26136654 PMCID: PMC4469831 DOI: 10.3389/fnins.2015.00220] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 06/02/2015] [Indexed: 12/27/2022] Open
Abstract
The brain noradrenergic system supplies the neurotransmitter norepinephrine throughout the brain via widespread efferent projections, and plays a pivotal role in modulating cognitive activities in the cortex. Profound noradrenergic degeneration in Alzheimer's disease (AD) patients has been observed for decades, with recent research suggesting that the locus coeruleus (where noradrenergic neurons are mainly located) is a predominant site where AD-related pathology begins. Mounting evidence indicates that the loss of noradrenergic innervation greatly exacerbates AD pathogenesis and progression, although the precise roles of noradrenergic components in AD pathogenesis remain unclear. The aim of this review is to summarize current findings on noradrenergic dysfunction in AD, as well as to point out deficiencies in our knowledge where more research is needed.
Collapse
Affiliation(s)
- Mary Gannon
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham Birmingham, AL, USA
| | - Pulin Che
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham Birmingham, AL, USA
| | - Yunjia Chen
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham Birmingham, AL, USA
| | - Kai Jiao
- Department of Genetics, University of Alabama at Birmingham Birmingham, AL, USA
| | - Erik D Roberson
- Department of Neurology, University of Alabama at Birmingham Birmingham, AL, USA
| | - Qin Wang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham Birmingham, AL, USA
| |
Collapse
|
22
|
Chang HS, Won ES, Lee HY, Ham BJ, Kim YG, Lee MS. Association of ARRB1 polymorphisms with the risk of major depressive disorder and with treatment response to mirtazapine. J Psychopharmacol 2015; 29:615-22. [PMID: 25294870 DOI: 10.1177/0269881114554273] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
β-Arrestin 1 is known to be involved in the pathophysiology of major depressive disorder (MDD) and in the underlying mechanism of action of antidepressant therapies. After we screened 39 ARRB1 polymorphisms, we investigated the associations of seven ARRB1 single-nucleotide polymorphisms (SNPs) with the risk of MDD in 270 patients with MDD and 204 normal subjects, and with mirtazapine treatment response in patients with MDD. The genotype distributions of -132C>T and IVS1+85T>C showed significant deviations from Hardy-Weinberg equilibrium in patients with MDD but not in normal subjects. After four and 12 weeks of mirtazapine treatment, the proportion of haplotype 1 (ht1) carriers was significantly higher in remitters than in non-remitters after corrections for multiple comparisons (corrected p=0.006 and 0.014 at four and 12 weeks, respectively). After eight and 12 weeks of treatment, scores on the 21-item Hamilton Depression Rating Scale (HAMD21) were significantly lower in patients with MDD with ARRB1 ht1 than in those without ht1. Similarly, after 8 and 12 weeks of treatment, the percent reduction in HAMD21 scores was significantly higher in patients with MDD with ARRB1 ht1 than in those without ht1. The ARRB1 polymorphisms represent promising genetic markers for the prediction of treatment responses to mirtazapine.
Collapse
Affiliation(s)
- Hun Soo Chang
- Department of Medical Bioscience, Soonchunhyang University, Bucheon, Republic of Korea
| | - Eun Soo Won
- Pharmacogenetic Research Center for Psychotropic Drugs, Korea University, Seoul, Korea Department of Psychiatry, College of Medicine, Korea University, Seoul, Korea
| | - Hwa-Young Lee
- Department of psychiatry, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Byung-Joo Ham
- Pharmacogenetic Research Center for Psychotropic Drugs, Korea University, Seoul, Korea Department of Psychiatry, College of Medicine, Korea University, Seoul, Korea
| | - Yong-Gu Kim
- Pharmacogenetic Research Center for Psychotropic Drugs, Korea University, Seoul, Korea Department of Psychiatry, College of Medicine, Korea University, Seoul, Korea
| | - Min-Soo Lee
- Pharmacogenetic Research Center for Psychotropic Drugs, Korea University, Seoul, Korea Department of Psychiatry, College of Medicine, Korea University, Seoul, Korea
| |
Collapse
|
23
|
Electroconvulsive shocks decrease α2-adrenoceptor binding in the Flinders rat model of depression. Eur Neuropsychopharmacol 2015; 25:404-12. [PMID: 25604421 DOI: 10.1016/j.euroneuro.2014.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 12/03/2014] [Accepted: 12/15/2014] [Indexed: 11/20/2022]
Abstract
Despite years of drug development, electroconvulsive therapy (ECT) remains the most effective treatment for severe depression. The exact therapeutic mechanism of action of ECT is still unresolved and therefore we tested the hypothesis that the beneficial effect of ECT could in part be the result of increased noradrenergic neurotransmission leading to a decrease in α2-adrenoceptor binding. We have previously shown that both the Flinders sensitive line (FSL) and Flinders resistant line (FRL) rats had altered α2-adrenoceptor binding compared to control Sprague-Dawley (SD) rats. In this study, we treated female FSL, FRL and SD rats with electroconvulsive shock (ECS), an animal model of ECT, or sham stimulation for 10 days before brains were removed and cut into 20µm thick sections. Densities of α2-adrenoceptors were measured by quantitative autoradiography in the hippocampus, thalamic nucleus, hypothalamus, amygdala, frontal cortex, insular cortex, and perirhinal cortex using the α2-adrenoceptor antagonist, [(3)H]RX 821002. ECS decreased the binding of α2-adrenoceptors in cortical regions in the FSL and cortical and amygdaloid regions in the control FRL rats compared to their respective sham treated group. The normal SD controls showed no significant response to ECS treatment. Our data suggest that the therapeutic effect of ECS may be mediated through a decrease of α2-adrenoceptors, probably due to a sustained increase in noradrenaline release. These data confirm the importance of the noradrenergic system and the α2-adrenoceptor in depression and in the mechanism of antidepressant treatments.
Collapse
|
24
|
Foster SR, Roura E, Molenaar P, Thomas WG. G protein-coupled receptors in cardiac biology: old and new receptors. Biophys Rev 2015; 7:77-89. [PMID: 28509979 DOI: 10.1007/s12551-014-0154-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 11/25/2014] [Indexed: 12/21/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are seven-transmembrane-spanning proteins that mediate cellular and physiological responses. They are critical for cardiovascular function and are targeted for the treatment of hypertension and heart failure. Nevertheless, current therapies only target a small fraction of the cardiac GPCR repertoire, indicating that there are many opportunities to investigate unappreciated aspects of heart biology. Here, we offer an update on the contemporary view of GPCRs and the complexities of their signalling, and review the roles of the 'classical' GPCRs in cardiovascular physiology and disease. We then provide insights into other GPCRs that have been less extensively studied in the heart, including orphan, odorant and taste receptors. We contend that these novel cardiac GPCRs contribute to heart function in health and disease and thereby offer exciting opportunities to therapeutically modulate heart function.
Collapse
Affiliation(s)
- Simon R Foster
- School of Biomedical Sciences, University of Queensland, St Lucia Campus, 4072, Brisbane, Australia
| | - Eugeni Roura
- School of Biomedical Sciences, University of Queensland, St Lucia Campus, 4072, Brisbane, Australia.,Centre for Nutrition & Food Sciences, Queensland Alliance for Agriculture and Food Innovation, University of Queensland, St Lucia Campus, Brisbane, Australia
| | - Peter Molenaar
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, St Lucia Campus, Brisbane, Australia.,School of Medicine, University of Queensland, St Lucia Campus, Brisbane, Australia
| | - Walter G Thomas
- School of Biomedical Sciences, University of Queensland, St Lucia Campus, 4072, Brisbane, Australia.
| |
Collapse
|
25
|
α2 Adrenergic Receptor Trafficking as a Therapeutic Target in Antidepressant Drug Action. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 132:207-25. [DOI: 10.1016/bs.pmbts.2015.03.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
26
|
Tricyclic antidepressants exhibit variable pharmacological profiles at the α(2A) adrenergic receptor. Biochem Biophys Res Commun 2014; 451:461-6. [PMID: 25128275 DOI: 10.1016/j.bbrc.2014.08.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 08/05/2014] [Indexed: 01/09/2023]
Abstract
Antidepressant mechanisms of action remain shrouded in mystery, greatly hindering our ability to develop therapeutics which can fully treat patients suffering from depressive disorders. In an attempt to shed new light on this topic, we have undertaken a series of studies investigating actions of tricyclic antidepressant drugs (TCAs) at the α2A adrenergic receptor (AR), a centrally important receptor, dysregulation of which has been linked to depression. Our previous work established a particular TCA, desipramine, as an arrestin-biased α2AAR ligand driving receptor endocytosis and downregulation but not canonical heterotrimeric G protein-mediated signaling. The present work is aimed at broadening our understanding of how members of the TCA drug class act at the α2AAR, as we have selected the closely related but subtly different TCAs imipramine and amitriptyline for evaluation. Our data demonstrate that these drugs do also function as direct arrestin-biased α2AAR ligands. However, these data reveal differences in receptor affinity and in the extent/nature of arrestin recruitment to and endocytosis of α2AARs. Specifically, amitriptyline exhibits an approximately 14-fold stronger interaction with the receptor, is a weaker driver of arrestin recruitment, and preferentially recruits a different arrestin subtype. Extent of endocytosis is similar for all TCAs studied so far, and occurs in an arrestin-dependent manner, although imipramine uniquely retains a slight ability to drive α2AAR endocytosis in arrestin-null cells. These findings signify an important expansion of our mechanistic understanding of antidepressant pharmacology, and provide useful insights for future medicinal chemistry efforts.
Collapse
|
27
|
Kurko D, Kapui Z, Nagy J, Lendvai B, Kolok S. Analysis of functional selectivity through G protein-dependent and -independent signaling pathways at the adrenergic α(2C) receptor. Brain Res Bull 2014; 107:89-101. [PMID: 25080296 DOI: 10.1016/j.brainresbull.2014.07.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/15/2014] [Accepted: 07/17/2014] [Indexed: 01/01/2023]
Abstract
Although G protein-coupled receptors (GPCRs) are traditionally categorized as Gs-, Gq-, or Gi/o-coupled, their signaling is regulated by multiple mechanisms. GPCRs can couple to several effector pathways, having the capacity to interact not only with more than one G protein subtype but also with alternative signaling or effector proteins such as arrestins. Moreover, GPCR ligands can have different efficacies for activating these signaling pathways, a characteristic referred to as biased agonism or functional selectivity. In this work our aim was to detect differences in the ability of various agonists acting at the α2C type of adrenergic receptors (α2C-ARs) to modulate cAMP accumulation, cytoplasmic Ca(2+) release, β-arrestin recruitment and receptor internalization. A detailed comparative pharmacological characterization of G protein-dependent and -independent signaling pathways was carried out using adrenergic agonists (norepinephrine, phenylephrine, brimonidine, BHT-920, oxymetazoline, clonidine, moxonidine, guanabenz) and antagonists (MK912, yohimbine). As initial analysis of agonist Emax and EC50 values suggested possible functional selectivity, ligand bias was quantified by applying the relative activity scale and was compared to that of the endogenous agonist norepinephrine. Values significantly different from 0 between pathways indicated an agonist that promoted different level of activation of diverse effector pathways most likely due to the stabilization of a subtly different receptor conformation from that induced by norepinephrine. Our results showed that a series of agonists acting at the α2C-AR displayed different degree of functional selectivity (bias factors ranging from 1.6 to 36.7) through four signaling pathways. As signaling via these pathways seems to have distinct functional and physiological outcomes, studying all these stages of receptor activation could have further implications for the development of more selective therapeutics with improved efficacy and/or fewer side effects.
Collapse
Affiliation(s)
- Dalma Kurko
- Pharmacological and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary.
| | - Zoltán Kapui
- Pharmacological and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - József Nagy
- Pharmacological and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - Balázs Lendvai
- Pharmacological and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - Sándor Kolok
- Pharmacological and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| |
Collapse
|
28
|
Luttrell LM. Minireview: More than just a hammer: ligand "bias" and pharmaceutical discovery. Mol Endocrinol 2014; 28:281-94. [PMID: 24433041 DOI: 10.1210/me.2013-1314] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Conventional orthosteric drug development programs targeting G protein-coupled receptors (GPCRs) have focused on the concepts of agonism and antagonism, in which receptor structure determines the nature of the downstream signal and ligand efficacy determines its intensity. Over the past decade, the emerging paradigms of "pluridimensional efficacy" and "functional selectivity" have revealed that GPCR signaling is not monolithic, and that ligand structure can "bias" signal output by stabilizing active receptor states in different proportions than the native ligand. Biased ligands are novel pharmacologic entities that possess the unique ability to qualitatively change GPCR signaling, in effect creating "new receptors" with distinct efficacy profiles driven by ligand structure. The promise of biased agonism lies in this ability to engender "mixed" effects not attainable using conventional agonists or antagonists, promoting therapeutically beneficial signals while antagonizing deleterious ones. Indeed, arrestin pathway-selective agonists for the type 1 parathyroid hormone and angiotensin AT1 receptors, and G protein pathway-selective agonists for the GPR109A nicotinic acid and μ-opioid receptors, have demonstrated unique, and potentially therapeutic, efficacy in cell-based assays and preclinical animal models. Conversely, activating GPCRs in "unnatural" ways may lead to downstream biological consequences that cannot be predicted from prior knowledge of the actions of the native ligand, especially in the case of ligands that selectively activate as-yet poorly characterized G protein-independent signaling networks mediated via arrestins. Although much needs to be done to realize the clinical potential of functional selectivity, biased GPCR ligands nonetheless appear to be important new additions to the pharmacologic toolbox.
Collapse
Affiliation(s)
- Louis M Luttrell
- Department of Medicine and Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425; and Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29401
| |
Collapse
|
29
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: G protein-coupled receptors. Br J Pharmacol 2013; 170:1459-581. [PMID: 24517644 PMCID: PMC3892287 DOI: 10.1111/bph.12445] [Citation(s) in RCA: 505] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. G protein-coupled receptors are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, catalytic receptors, nuclear hormone receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|
30
|
Dexamethasone in the presence of desipramine enhances MAPK/ERK1/2 signaling possibly via its interference with β-arrestin. J Neural Transm (Vienna) 2013; 121:289-98. [PMID: 24132698 DOI: 10.1007/s00702-013-1099-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 10/05/2013] [Indexed: 01/07/2023]
Abstract
Antidepressant medication is the standard treatment for major depression disorder (MDD). However, the response to these treatments is often incomplete and many patients remain refractory. In the present study, we show that the glucocorticoid receptor (GR) agonist dexamethasone (DEX) increased MAPK/ERK1/2 signaling in the presence of the noradrenergic antidepressant, desipramine (DMI), while no such effect was induced by DEX or DMI alone in human neuroblastoma SH-SY5Y cells. This enhancement was dependent on the activation of both α(2) adrenergic receptors (AR) and GR. The timing of MAPK/ERK1/2 activation as well as DEX-induced reduction in membranous α(2) AR suggests the involvement of a β-arrestin-dependent mechanism. In line with the latter, DEX increased cytosolic and decreased membranous levels of β-arrestin. Concomitantly, DEX induced a time-dependent increase in cytosolic α(2) AR-β-arrestin interaction and a decrease in β-arrestin interaction with Mdm2 E3 ubiquitin ligase. All of these effects of DEX were prevented by the GR antagonist RU486. Our data suggest an additional intracellular role for DEX, in which activation of GR interferes with the trafficking and degradation of β-arrestin-α2c-AR complex. We suggest that such an interaction in the presence of DMI can enhance MAPK/ERK1/2 signaling, a key player in neural plasticity and neurogenesis processes, which is impaired in MDD, while stimulated by antidepressants.
Collapse
|
31
|
Cottingham C, Lu R, Jiao K, Wang Q. Cross-talk from β-adrenergic receptors modulates α2A-adrenergic receptor endocytosis in sympathetic neurons via protein kinase A and spinophilin. J Biol Chem 2013; 288:29193-205. [PMID: 23965992 DOI: 10.1074/jbc.m113.469494] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Inter-regulation of adrenergic receptors (ARs) via cross-talk is a long appreciated but mechanistically unclear physiological phenomenon. Evidence from the AR literature and our own extensive studies on regulation of α2AARs by the scaffolding protein spinophilin have illuminated a potential novel mechanism for cross-talk from β to α2ARs. In the present study, we have characterized a mode of endogenous AR cross-talk in native adrenergic neurons whereby canonical βAR-mediated signaling modulates spinophilin-regulated α2AAR endocytosis through PKA. Our findings demonstrate that co-activation of β and α2AARs, either by application of endogenous agonist or by simultaneous stimulation with distinct selective agonists, results in acceleration of endogenous α2AAR endocytosis in native neurons. We show that receptor-independent PKA activation by forskolin is sufficient to accelerate α2AAR endocytosis and that α2AAR stimulation alone drives accelerated endocytosis in spinophilin-null neurons. Endocytic response acceleration by β/α2AAR co-activation is blocked by PKA inhibition and lost in spinophilin-null neurons, consistent with our previous finding that spinophilin is a substrate for phosphorylation by PKA that disrupts its interaction with α2AARs. Importantly, we show that α2AR agonist-mediated α2AAR/spinophilin interaction is blocked by βAR co-activation in a PKA-dependent fashion. We therefore propose a novel mechanism for cross-talk from β to α2ARs, whereby canonical βAR-mediated signaling coupled to PKA activation results in phosphorylation of spinophilin, disrupting its interaction with α2AARs and accelerating α2AAR endocytic responses. This mechanism of cross-talk has significant implications for endogenous adrenergic physiology and for therapeutic targeting of β and α2AARs.
Collapse
|
32
|
Effect of acute stressor and serotonin transporter genotype on amygdala first wave transcriptome in mice. PLoS One 2013; 8:e58880. [PMID: 23536833 PMCID: PMC3594195 DOI: 10.1371/journal.pone.0058880] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 02/07/2013] [Indexed: 12/17/2022] Open
Abstract
The most prominent brain region evaluating the significance of external stimuli immediately after their onset is the amygdala. Stimuli evaluated as being stressful actuate a number of physiological processes as an immediate stress response. Variation in the serotonin transporter gene has been associated with increased anxiety- and depression-like behavior, altered stress reactivity and adaptation, and pathophysiology of stress-related disorders. In this study the instant reactions to an acute stressor were measured in a serotonin transporter knockout mouse model. Mice lacking the serotonin transporter were verified to be more anxious than their wild-type conspecifics. Genome-wide gene expression changes in the amygdala were measured after the mice were subjected to control condition or to an acute stressor of one minute exposure to water. The dissection of amygdalae and stabilization of RNA was conducted within nine minutes after the onset of the stressor. This extremely short protocol allowed for analysis of first wave primary response genes, typically induced within five to ten minutes of stimulation, and was performed using Affymetrix GeneChip Mouse Gene 1.0 ST Arrays. RNA profiling revealed a largely new set of differentially expressed primary response genes between the conditions acute stress and control that differed distinctly between wild-type and knockout mice. Consequently, functional categorization and pathway analysis indicated genes related to neuroplasticity and adaptation in wild-types whereas knockouts were characterized by impaired plasticity and genes more related to chronic stress and pathophysiology. Our study therefore disclosed different coping styles dependent on serotonin transporter genotype even directly after the onset of stress and accentuates the role of the serotonergic system in processing stressors and threat in the amygdala. Moreover, several of the first wave primary response genes that we found might provide promising targets for future therapeutic interventions of stress-related disorders also in humans.
Collapse
|
33
|
Kenakin T. New concepts in pharmacological efficacy at 7TM receptors: IUPHAR review 2. Br J Pharmacol 2013; 168:554-75. [PMID: 22994528 PMCID: PMC3579279 DOI: 10.1111/j.1476-5381.2012.02223.x] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 08/03/2012] [Accepted: 09/12/2012] [Indexed: 01/14/2023] Open
Abstract
The present-day concept of drug efficacy has changed completely from its original description as the property of agonists that causes tissue activation. The ability to visualize the multiple behaviours of seven transmembrane receptors has shown that drugs can have many efficacies and also that the transduction of drug stimulus to various cellular stimulus-response cascades can be biased towards some but not all pathways. This latter effect leads to agonist 'functional selectivity', which can be favourable for the improvement of agonist therapeutics. However, in addition, biased agonist potency becomes cell type dependent with the loss of the monotonic behaviour of stimulus-response mechanisms, leading to potential problems in agonist quantification. This has an extremely important effect on the discovery process for new agonists since it now cannot be assumed that a given screening or lead optimization assay will correctly predict therapeutic behaviour. This review discusses these ideas and how new approaches to quantifying agonist effect may be used to circumvent the cell type dependence of agonism. This article, written by a corresponding member of the International Union of Basic and Clinical Pharmacology Committee on Receptor Nomenclature and Drug Classification (NC-IUPHAR), reviews our current understanding of the interaction of ligands with seven transmembrane receptors. Further information on these pharmacological concepts is being incorporated into the IUPHAR/BPS database GuideToPharmacology.org.
Collapse
Affiliation(s)
- Terry Kenakin
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
34
|
β-Arrestins in the Central Nervous System. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 118:267-95. [DOI: 10.1016/b978-0-12-394440-5.00011-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
35
|
Systems Analysis of Arrestin Pathway Functions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 118:431-67. [DOI: 10.1016/b978-0-12-394440-5.00017-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
36
|
Cottingham C, Wang Q. α2 adrenergic receptor dysregulation in depressive disorders: implications for the neurobiology of depression and antidepressant therapy. Neurosci Biobehav Rev 2012; 36:2214-25. [PMID: 22910678 DOI: 10.1016/j.neubiorev.2012.07.011] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 06/27/2012] [Accepted: 07/25/2012] [Indexed: 12/25/2022]
Abstract
Dysfunction in noradrenergic neurotransmission has long been theorized to occur in depressive disorders. The α2 adrenergic receptor (AR) family, as a group of key players in regulating the noradrenergic system, has been investigated for involvement in the neurobiology of depression and mechanisms of antidepressant therapies. However, a clear picture of the α2ARs in depressive disorders has not been established due to the existence of apparently conflicting findings in the literature. In this article, we report that a careful accounting of methodological differences within the literature can resolve the present lack of consensus on involvement of α2ARs in depression. In particular, the pharmacological properties of the radioligand (e.g. agonist versus antagonist) utilized for determining receptor density are crucial in determining study outcome. Upregulation of α2AR density detected by radiolabeled agonists but not by antagonists in patients with depressive disorders suggests a selective increase in the density of high-affinity conformational state α2ARs, which is indicative of enhanced G protein coupling to the receptor. Importantly, this high-affinity state α2AR upregulation can be normalized with antidepressant treatments. Thus, depressive disorders appear to be associated with increased α2AR sensitivity and responsiveness, which may represent a physiological basis for the putative noradrenergic dysfunction in depressive disorders. In addition, we review changes in some key α2AR accessory proteins in depressive disorders and discuss their potential contribution to α2AR dysfunction.
Collapse
Affiliation(s)
- Christopher Cottingham
- Department of Cell, Developmental & Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | |
Collapse
|
37
|
Kenakin T. Casting a wider net: whole-cell assays to capture varied and biased signaling. Mol Pharmacol 2012; 82:571-4. [PMID: 22828801 DOI: 10.1124/mol.112.081117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The observation of complex receptor behaviors has shown how ligands can have multiple efficacies and can also differentially stimulate certain cellular signaling pathways over others (i.e., biased signaling). Conventional pharmacological assays (usually proximal to the receptor) will detect ligands that produce the signal defined by the assay (i.e., Ca²⁺, cAMP, and others) but otherwise may miss biased ligands that produce little activation of pathways not measured by the assay. In theory, this is less of a hazard for generic whole-cell assays, which may be sensitive to multiple signaling inputs. Whole-cell assays have the advantage of detecting effects induced by a variety of receptor interactions with cytosolic proteins, including those that may be previously unknown. These ideas are discussed within the context of the high-throughput flow cytometry measurement of receptor internalization described by Wu et al. in the current issue of the journal. The internalization of receptors can be a useful therapeutic modality and the article by Wu et al. illustrates how this new assay, targeted to downstream cellular effects, can uncover unique ligand efficacies linked to receptor internalization.
Collapse
Affiliation(s)
- Terry Kenakin
- Department of Pharmacology, University of North Carolina School of Medicine, 120 Mason Farm Rd., Room 4042 Genetic Medicine Building, CB#7365, Chapel Hill, NC 27599-7365, USA.
| |
Collapse
|
38
|
Yang J, Dolinger M, Ritaccio G, Mazurkiewicz J, Conti D, Zhu X, Huang Y. Leucine stimulates insulin secretion via down-regulation of surface expression of adrenergic α2A receptor through the mTOR (mammalian target of rapamycin) pathway: implication in new-onset diabetes in renal transplantation. J Biol Chem 2012; 287:24795-806. [PMID: 22645144 DOI: 10.1074/jbc.m112.344259] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The amino acid leucine is a potent secretagogue, capable of inducing insulin secretion. It also plays an important role in the regulation of mTOR activity, therefore, providing impetus to investigate if a leucine-sensing mechanism in the mTOR pathway is involved in insulin secretion. We found that leucine-induced insulin secretion was inhibited by both the mTOR inhibitor rapamycin as well as the adrenergic α2 receptor agonist clonidine. We also demonstrated that leucine down-regulated the surface expression of adrenergic α2A receptor via activation of the mTOR pathway. The leucine stimulatory effect on insulin secretion was attenuated in diabetic Goto-Kakizaki rats that overexpress adrenergic α2A receptors, confirming the role of leucine in insulin secretion. Thus, our data demonstrate that leucine regulates insulin secretion by modulating adrenergic α2 receptors through the mTOR pathway. The role of the mTOR pathway in metabolic homeostasis led us to a second important finding in this study; retrospective analysis of clinical data showed that co-administration of rapamycin and clonidine was associated with an increased incidence of new-onset diabetes in renal transplantation patients over those receiving rapamycin alone. We believe that inhibition of mTOR by rapamycin along with activation of adrenergic α2 receptors by clonidine represents a double-hit to pancreatic islets that synergistically disturbs glucose homeostasis. This new insight may have important implications for the clinical management of renal transplant patients.
Collapse
Affiliation(s)
- Jun Yang
- Center for Neuropharmacology and Neuroscience, Albany Medical College, Albany, New York 12208, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Neurabin scaffolding of adenosine receptor and RGS4 regulates anti-seizure effect of endogenous adenosine. J Neurosci 2012; 32:2683-95. [PMID: 22357852 DOI: 10.1523/jneurosci.4125-11.2011] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Endogenous adenosine is an essential protective agent against neural damage by various insults to the brain. However, the therapeutic potential of adenosine receptor-directed ligands for neuroprotection is offset by side effects in peripheral tissues and organs. An increase in adenosine receptor responsiveness to endogenous adenosine would enhance neuroprotection while avoiding the confounding effects of exogenous ligands. Here we report novel regulation of adenosine-evoked responses by a neural tissue-specific protein, neurabin. Neurabin attenuated adenosine A(1) receptor (A1R) signaling by assembling a complex between the A1R and the regulator of G-protein signaling 4 (RGS4), a protein known to turn off G-protein signaling. Inactivation of the neurabin gene enhanced A1R signaling and promoted the protective effect of adenosine against excitotoxic seizure and neuronal death in mice. Furthermore, administration of a small molecule inhibitor of RGS4 significantly attenuated seizure severity in mice. Notably, the dose of kainate capable of inducing an ∼50% rate of death in wild-type (WT) mice did not affect neurabin-null mice or WT mice cotreated with an RGS4 inhibitor. The enhanced anti-seizure and neuroprotective effect achieved by disruption of the A1R/neurabin/RGS4 complex is elicited by the on-site and on-demand release of endogenous adenosine, and does not require administration of A1R ligands. These data identify neurabin-RGS4 as a novel tissue-selective regulatory mechanism for fine-tuning adenosine receptor function in the nervous system. Moreover, these findings implicate the A1R/neurabin/RGS4 complex as a valid therapeutic target for specifically manipulating the neuroprotective effects of endogenous adenosine.
Collapse
|
40
|
Cottingham C, Jones A, Wang Q. Desipramine selectively potentiates norepinephrine-elicited ERK1/2 activation through the α2A adrenergic receptor. Biochem Biophys Res Commun 2012; 420:161-5. [PMID: 22405824 DOI: 10.1016/j.bbrc.2012.02.135] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 02/24/2012] [Indexed: 10/28/2022]
Abstract
The precise physiological effects of antidepressant drugs, and in particular their actions at non-monoamine transporter targets, are largely unknown. We have recently identified the tricyclic antidepressant drug desipramine (DMI) as a direct ligand at the α(2A) adrenergic receptor (AR) without itself driving heterotrimeric G protein/downstream effector activation [5]. In this study, we report our novel finding that DMI modulates α(2A)AR signaling in response to the endogenous agonist norepinephrine (NE). DMI acted as a signaling potentiator, selectively enhancing NE-induced α(2A)AR-mediated ERK1/2 MAPK signaling. This potentiation of ERK1/2 activation was observed as an increase in NE response sensitivity and a prolongation of the activation kinetics. DMI in a physiologically relevant ratio with NE effectively turned on ERK1/2 signaling that is lacking in response to physiological NE alone. Further, the DMI-induced ERK1/2 potentiation relied on heterotrimeric G(i/o) proteins and was arrestin-independent. This modulatory effect of DMI on NE signaling provides novel insight into the effects of this antidepressant drug on the noradrenergic system which it regulates, insight which enhances our understanding of the therapeutic mechanism for DMI.
Collapse
Affiliation(s)
- Christopher Cottingham
- Department of Physiology & Biophysics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | |
Collapse
|
41
|
Cottingham C, Li X, Wang Q. Noradrenergic antidepressant responses to desipramine in vivo are reciprocally regulated by arrestin3 and spinophilin. Neuropharmacology 2012; 62:2354-62. [PMID: 22369787 DOI: 10.1016/j.neuropharm.2012.02.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 02/09/2012] [Accepted: 02/12/2012] [Indexed: 01/21/2023]
Abstract
Many antidepressant drugs, including the tricyclic antidepressant desipramine (DMI), are broadly understood to function by modulating central noradrenergic neurotransmission. α(2) adrenergic receptors (α(2)ARs) are key regulators of the noradrenergic system, and previous work has implicated α(2)ARs in mediating the antidepressant activity of DMI in the rodent forced swim test (FST). However, little is known about intracellular regulators of antidepressant drug action. α(2)AR function is tightly regulated by its intracellular interacting partners arrestin and the dendritic protein spinophilin. We have previously established the competitive and reciprocal nature of these interacting proteins at the α(2)AR in the context of classic agonist effects, and have shown DMI to be a direct arrestin-biased ligand at the receptor. In the present study, we report that mice deficient in the α(2A)AR subtype lack DMI-induced antidepressant behavioral effects in the FST. As well, mice deficient in arrestin3 lack antidepressant response to DMI, while spinophilin-null mice have enhanced antidepressant response to DMI compared with wild-type controls, indicating that this α(2A)AR-mediated response is reciprocally regulated by arrestin and spinophilin. The characteristic of α(2A)AR-dependence and arrestin3 involvement was shared by the antidepressant effect of the classic α(2)AR agonist clonidine but not the non-tricyclic norepinephrine reuptake inhibitor reboxetine, supporting a model whereby DMI exerts its antidepressant effect through direct engagement of the α(2A)AR and arrestin3. Our results implicate arrestin- and spinophilin-mediated regulation of the α(2A)AR in the pharmacology of the noradrenergic antidepressant DMI, and suggest that manipulation of this mode of receptor regulation may represent a novel and viable therapeutic strategy.
Collapse
Affiliation(s)
- Christopher Cottingham
- Department of Physiology & Biophysics, University of Alabama at Birmingham, 986 MCLM 1918 University Blvd., Birmingham, AL 35294, USA
| | | | | |
Collapse
|
42
|
Signalling bias in new drug discovery: detection, quantification and therapeutic impact. Nat Rev Drug Discov 2012; 12:205-16. [PMID: 23411724 DOI: 10.1038/nrd3954] [Citation(s) in RCA: 585] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Agonists of seven-transmembrane receptors, also known as G protein-coupled receptors (GPCRs), do not uniformly activate all cellular signalling pathways linked to a given seven-transmembrane receptor (a phenomenon termed ligand or agonist bias); this discovery has changed how high-throughput screens are designed and how lead compounds are optimized for therapeutic activity. The ability to experimentally detect ligand bias has necessitated the development of methods for quantifying agonist bias in a way that can be used to guide structure-activity studies and the selection of drug candidates. Here, we provide a viewpoint on which methods are appropriate for quantifying bias, based on knowledge of how cellular and intracellular signalling proteins control the conformation of seven-transmembrane receptors. We also discuss possible predictions of how biased molecules may perform in vivo, and what potential therapeutic advantages they may provide.
Collapse
|