1
|
Peeters LD, Wills LJ, Cuozzo AM, Ivanich KL, Turney SE, Bullock LP, Price RM, Gass JT, Brown RW. Modulation of mGlu5 reduces rewarding associative properties of nicotine via changes in mesolimbic plasticity: Relevance to comorbid cigarette smoking in psychosis. Pharmacol Biochem Behav 2024; 239:173752. [PMID: 38521210 PMCID: PMC11088493 DOI: 10.1016/j.pbb.2024.173752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/13/2024] [Accepted: 03/20/2024] [Indexed: 03/25/2024]
Abstract
RATIONALE Antipsychotic medications that are used to treat psychosis are often limited in their efficacy by high rates of severe side effects. Treatment success in schizophrenia is further complicated by high rates of comorbid nicotine use. Dopamine D2 heteroreceptor complexes have recently emerged as targets for the development of more efficacious pharmaceutical treatments for schizophrenia. OBJECTIVE The current study sought to explore the use of the positive allosteric modulator of the mGlu5 receptor 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB) as a treatment to reduce symptoms related to psychosis and comorbid nicotine use. METHODS Neonatal treatment of animals with the dopamine D2-like receptor agonist quinpirole (NQ) from postnatal day (P)1-21 produces a lifelong increase in D2 receptor sensitivity, showing relevance to psychosis and comorbid tobacco use disorder. Following an 8-day conditioning paradigm, brain tissue in the mesolimbic pathway was analyzed for several plasticity markers, including brain derived neurotrophic factor (BDNF), phosphorylated p70 ribosomal S6 kinase (phospho-p70S6K), and cadherin-13 (Cdh13). RESULTS Pretreatment with CDPPB was effective to block enhanced nicotine conditioned place preference observed in NQ-treated animals. Pretreatment was additionally effective to block the nicotine-induced increase in BDNF and sex-dependent increases in cadherin-13 in the ventral tegmental area (VTA), as well as increased phospho-p70S6K in the nucleus accumbens (NAcc) shell found in NQ-treated animals. CONCLUSION In conjunction with prior work, the current study suggests positive allosteric modulation of the mGlu5 receptor, an emerging target for schizophrenia therapeutics, may be effective for the treatment of comorbid nicotine abuse in psychosis.
Collapse
Affiliation(s)
- Loren D Peeters
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States of America
| | - Liza J Wills
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States of America
| | - Anthony M Cuozzo
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States of America
| | - Kira L Ivanich
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States of America
| | - Seth E Turney
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States of America
| | - Luke P Bullock
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States of America
| | - Robert M Price
- Department of Mathematics and Statistics, East Tennessee State University, Johnson City, TN 37614, United States of America
| | - Justin T Gass
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States of America
| | - Russell W Brown
- Department of Biomedical Sciences, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, United States of America.
| |
Collapse
|
2
|
Guidolin D, Tortorella C, Marcoli M, Cervetto C, De Caro R, Maura G, Agnati LF. Modulation of Neuron and Astrocyte Dopamine Receptors via Receptor-Receptor Interactions. Pharmaceuticals (Basel) 2023; 16:1427. [PMID: 37895898 PMCID: PMC10610355 DOI: 10.3390/ph16101427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 09/29/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Dopamine neurotransmission plays critical roles in regulating complex cognitive and behavioral processes including reward, motivation, reinforcement learning, and movement. Dopamine receptors are classified into five subtypes, widely distributed across the brain, including regions responsible for motor functions and specific areas related to cognitive and emotional functions. Dopamine also acts on astrocytes, which express dopamine receptors as well. The discovery of direct receptor-receptor interactions, leading to the formation of multimeric receptor complexes at the cell membrane and providing the cell decoding apparatus with flexible dynamics in terms of recognition and signal transduction, has expanded the knowledge of the G-protein-coupled receptor-mediated signaling processes. The purpose of this review article is to provide an overview of currently identified receptor complexes containing dopamine receptors and of their modulatory action on dopamine-mediated signaling between neurons and between neurons and astrocytes. Pharmacological possibilities offered by targeting receptor complexes in terms of addressing neuropsychiatric disorders associated with altered dopamine signaling will also be briefly discussed.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Neuroscience, University of Padova, 35122 Padova, Italy; (C.T.); (R.D.C.)
| | - Cinzia Tortorella
- Department of Neuroscience, University of Padova, 35122 Padova, Italy; (C.T.); (R.D.C.)
| | - Manuela Marcoli
- Department of Pharmacy, University of Genova, 16126 Genova, Italy; (M.M.); (C.C.); (G.M.)
| | - Chiara Cervetto
- Department of Pharmacy, University of Genova, 16126 Genova, Italy; (M.M.); (C.C.); (G.M.)
| | - Raffaele De Caro
- Department of Neuroscience, University of Padova, 35122 Padova, Italy; (C.T.); (R.D.C.)
| | - Guido Maura
- Department of Pharmacy, University of Genova, 16126 Genova, Italy; (M.M.); (C.C.); (G.M.)
| | - Luigi F. Agnati
- Department of Biomedical, Metabolic Sciences and Neuroscience, University of Modena and Reggio Emilia, 41121 Modena, Italy;
| |
Collapse
|
3
|
Dopamine Dynamics and Neurobiology of Non-Response to Antipsychotics, Relevance for Treatment Resistant Schizophrenia: A Systematic Review and Critical Appraisal. Biomedicines 2023; 11:biomedicines11030895. [PMID: 36979877 PMCID: PMC10046109 DOI: 10.3390/biomedicines11030895] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/08/2023] [Accepted: 03/12/2023] [Indexed: 03/17/2023] Open
Abstract
Treatment resistant schizophrenia (TRS) is characterized by a lack of, or suboptimal response to, antipsychotic agents. The biological underpinnings of this clinical condition are still scarcely understood. Since all antipsychotics block dopamine D2 receptors (D2R), dopamine-related mechanisms should be considered the main candidates in the neurobiology of antipsychotic non-response, although other neurotransmitter systems play a role. The aims of this review are: (i) to recapitulate and critically appraise the relevant literature on dopamine-related mechanisms of TRS; (ii) to discuss the methodological limitations of the studies so far conducted and delineate a theoretical framework on dopamine mechanisms of TRS; and (iii) to highlight future perspectives of research and unmet needs. Dopamine-related neurobiological mechanisms of TRS may be multiple and putatively subdivided into three biological points: (1) D2R-related, including increased D2R levels; increased density of D2Rs in the high-affinity state; aberrant D2R dimer or heteromer formation; imbalance between D2R short and long variants; extrastriatal D2Rs; (2) presynaptic dopamine, including low or normal dopamine synthesis and/or release compared to responder patients; and (3) exaggerated postsynaptic D2R-mediated neurotransmission. Future points to be addressed are: (i) a more neurobiologically-oriented phenotypic categorization of TRS; (ii) implementation of neurobiological studies by directly comparing treatment resistant vs. treatment responder patients; (iii) development of a reliable animal model of non-response to antipsychotics.
Collapse
|
4
|
Prasad K, de Vries EFJ, Sijbesma JW, Garcia-Varela L, Vazquez-Matias DA, Moraga-Amaro R, Willemsen ATM, Dierckx RAJO, van Waarde A. Impact of an Adenosine A 2A Receptor Agonist and Antagonist on Binding of the Dopamine D 2 Receptor Ligand [ 11C]raclopride in the Rodent Striatum. Mol Pharm 2022; 19:2992-3001. [PMID: 35849844 PMCID: PMC9346611 DOI: 10.1021/acs.molpharmaceut.2c00450] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/29/2022] [Accepted: 07/01/2022] [Indexed: 11/29/2022]
Abstract
Adenosine A2A and dopamine D2 receptors in the basal ganglia form heterotetrameric structures that are involved in the regulation of motor activity and neuropsychiatric functions. The present study examines the A2A receptor-mediated modulation of D2 receptor binding in vivo using positron emission tomography (PET) with the D2 antagonist tracer [11C]raclopride. Healthy male Wistar rats (n = 8) were scanned (60 min dynamic scan) with [11C]raclopride at baseline and 7 days later following an acute administration of the A2A agonist CGS21680 (1 mg/kg), using a MicroPET Focus-220 camera. Nondisplaceable binding potential (BPND) values were calculated using a simplified reference tissue model (SRTM), with cerebellum as the reference tissue. SRTM analysis did not show any significant changes in [11C]raclopride BPND (p = 0.102) in striatum after CGS21680 administration compared to the baseline. As CGS21680 strongly affects hemodynamics, we also used arterial blood sampling and a metabolite-corrected plasma input function for compartment modeling using the reversible two-tissue compartment model (2TCM) to obtain the BPND from the k3/k4 ratio and from the striatum/cerebellum volume of distribution ratio (DVR) in a second group of animals. These rats underwent dynamic [11C]raclopride scans after pretreatment with a vehicle (n = 5), a single dose of CGS21680 (1 mg/kg, n = 5), or a single dose of the A2A antagonist KW6002 (1 mg/kg, n = 5). The parent fraction in plasma was significantly higher in the CGS21680-treated group (p = 0.0001) compared to the vehicle-treated group. GCS21680 administration significantly reduced the striatal k3/k4 ratio (p < 0.01), but k3 and k4 estimates may be less reliable. The BPND (DVR-1) decreased from 1.963 ± 0.27 in the vehicle-treated group to 1.53 ± 0.55 (p = 0.080) or 1.961 ± 0.11 (p = 0.993) after the administration of CGS21680 or KW6002, respectively. Our study suggests that the A2A agonist CGS21680, but not the antagonist KW6002, may reduce the D2 receptor availability in the striatum.
Collapse
Affiliation(s)
- Kavya Prasad
- Department of Nuclear Medicine
and Molecular Imaging, University Medical
Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box 30001, 9713 GZ Groningen, The Netherlands
| | - Erik F. J. de Vries
- Department of Nuclear Medicine
and Molecular Imaging, University Medical
Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box 30001, 9713 GZ Groningen, The Netherlands
| | - Jürgen W.
A. Sijbesma
- Department of Nuclear Medicine
and Molecular Imaging, University Medical
Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box 30001, 9713 GZ Groningen, The Netherlands
| | - Lara Garcia-Varela
- Department of Nuclear Medicine
and Molecular Imaging, University Medical
Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box 30001, 9713 GZ Groningen, The Netherlands
| | - Daniel A. Vazquez-Matias
- Department of Nuclear Medicine
and Molecular Imaging, University Medical
Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box 30001, 9713 GZ Groningen, The Netherlands
| | - Rodrigo Moraga-Amaro
- Department of Nuclear Medicine
and Molecular Imaging, University Medical
Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box 30001, 9713 GZ Groningen, The Netherlands
| | - Antoon T. M. Willemsen
- Department of Nuclear Medicine
and Molecular Imaging, University Medical
Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box 30001, 9713 GZ Groningen, The Netherlands
| | - Rudi A. J. O. Dierckx
- Department of Nuclear Medicine
and Molecular Imaging, University Medical
Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box 30001, 9713 GZ Groningen, The Netherlands
| | - Aren van Waarde
- Department of Nuclear Medicine
and Molecular Imaging, University Medical
Center Groningen, University of Groningen, Hanzeplein 1, P.O. Box 30001, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
5
|
Dysfunctional Heteroreceptor Complexes as Novel Targets for the Treatment of Major Depressive and Anxiety Disorders. Cells 2022; 11:cells11111826. [PMID: 35681521 PMCID: PMC9180493 DOI: 10.3390/cells11111826] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/10/2022] [Accepted: 05/20/2022] [Indexed: 02/01/2023] Open
Abstract
Among mental diseases, major depressive disorder (MDD) and anxiety deserve a special place due to their high prevalence and their negative impact both on society and patients suffering from these disorders. Consequently, the development of novel strategies designed to treat them quickly and efficiently, without or at least having limited side effects, is considered a highly important goal. Growing evidence indicates that emerging properties are developed on recognition, trafficking, and signaling of G-protein coupled receptors (GPCRs) upon their heteromerization with other types of GPCRs, receptor tyrosine kinases, and ionotropic receptors such as N-methyl-D-aspartate (NMDA) receptors. Therefore, to develop new treatments for MDD and anxiety, it will be important to identify the most vulnerable heteroreceptor complexes involved in MDD and anxiety. This review focuses on how GPCRs, especially serotonin, dopamine, galanin, and opioid heteroreceptor complexes, modulate synaptic and volume transmission in the limbic networks of the brain. We attempt to provide information showing how these emerging concepts can contribute to finding new ways to treat both MDD and anxiety disorders.
Collapse
|
6
|
Adenosine Receptors in Neuropsychiatric Disorders: Fine Regulators of Neurotransmission and Potential Therapeutic Targets. Int J Mol Sci 2022; 23:ijms23031219. [PMID: 35163142 PMCID: PMC8835915 DOI: 10.3390/ijms23031219] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 12/16/2022] Open
Abstract
Adenosine exerts an important role in the modulation of central nervous system (CNS) activity. Through the interaction with four G-protein coupled receptor (GPCR) subtypes, adenosine subtly regulates neurotransmission, interfering with the dopaminergic, glutamatergic, noradrenergic, serotoninergic, and endocannabinoid systems. The inhibitory and facilitating actions of adenosine on neurotransmission are mainly mediated by A1 and A2A adenosine receptors (ARs), respectively. Given their role in the CNS, ARs are promising therapeutic targets for neuropsychiatric disorders where altered neurotransmission represents the most likely etiological hypothesis. Activating or blocking ARs with specific pharmacological agents could therefore restore the balance of altered neurotransmitter systems, providing the rationale for the potential treatment of these highly debilitating conditions. In this review, we summarize and discuss the most relevant studies concerning AR modulation in psychotic and mood disorders such as schizophrenia, bipolar disorders, depression, and anxiety, as well as neurodevelopment disorders such as autism spectrum disorder (ASD), fragile X syndrome (FXS), attention-deficit hyperactivity disorder (ADHD), and neuropsychiatric aspects of neurodegenerative disorders.
Collapse
|
7
|
Brown RW, Varnum CG, Wills LJ, Peeters LD, Gass JT. Modulation of mGlu5 improves sensorimotor gating deficits in rats neonatally treated with quinpirole through changes in dopamine D2 signaling. Pharmacol Biochem Behav 2021; 211:173292. [PMID: 34710401 PMCID: PMC9176413 DOI: 10.1016/j.pbb.2021.173292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/19/2022]
Abstract
This study analyzed whether the positive allosteric modulator of metabotropic glutamate receptor type 5 (mGlu5) 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl) benzamide (CDPPB) would alleviate deficits in prepulse inhibition (PPI) and affect dopamine (DA) D2 signaling in the dorsal striatum and prefrontal cortex (PFC) in the neonatal quinpirole (NQ) model of schizophrenia (SZ). Male and female Sprague-Dawley rats were neonatally treated with either saline (NS) or quinpirole HCL (1 mg/kg; NQ), a DAD2 receptor agonist, from postnatal days (P) 1-21. Rats were raised to P44 and behaviorally tested on PPI from P44-P48. Before each trial, rats were subcutaneous (sc) administered saline or CDPPB (10 mg/kg or 30 mg/kg). On P50, rats were given a spontaneous locomotor activity test after CDPPB or saline administration. On P51, the dorsal striatum and PFC were evaluated for both arrestin-2 (βA-2) and phospho-AKT protein levels. NQ-treated rats demonstrated a significant deficit in PPI, which was alleviated to control levels by the 30 mg/kg dose of CDPPB. There were no significant effects of CDPPB on locomotor activity. NQ treatment increased βA-2 and decreased phospho-AKT in both the dorsal striatum and PFC, consistent with an increase DAD2 signaling. The 30 mg/kg dose of CDPPB significantly reversed changes in βA-2 in the dorsal striatum and PFC and phospho-AKT in the PFC equivalent to controls. Both doses of CDPPB produced a decrease of phospho-AKT in the PFC compared to controls. This study revealed that a mGlu5 positive allosteric modulator was effective to alleviate PPI deficits and striatal DAD2 signaling in the NQ model of SZ.
Collapse
Affiliation(s)
- Russell W Brown
- Department of Biomedical Sciences James H. Quillen College of Medicine, East Tennessee State University Johnson City, TN 37614, United States of America.
| | - Christopher G Varnum
- Department of Biomedical Sciences James H. Quillen College of Medicine, East Tennessee State University Johnson City, TN 37614, United States of America
| | - Liza J Wills
- Department of Biomedical Sciences James H. Quillen College of Medicine, East Tennessee State University Johnson City, TN 37614, United States of America
| | - Loren D Peeters
- Department of Biomedical Sciences James H. Quillen College of Medicine, East Tennessee State University Johnson City, TN 37614, United States of America
| | - Justin T Gass
- Department of Biomedical Sciences James H. Quillen College of Medicine, East Tennessee State University Johnson City, TN 37614, United States of America
| |
Collapse
|
8
|
Serotonin Heteroreceptor Complexes and Their Integration of Signals in Neurons and Astroglia-Relevance for Mental Diseases. Cells 2021; 10:cells10081902. [PMID: 34440670 PMCID: PMC8392445 DOI: 10.3390/cells10081902] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/16/2021] [Accepted: 07/23/2021] [Indexed: 12/28/2022] Open
Abstract
The heteroreceptor complexes present a novel biological principle for signal integration. These complexes and their allosteric receptor-receptor interactions are bidirectional and novel targets for treatment of CNS diseases including mental diseases. The existence of D2R-5-HT2AR heterocomplexes can help explain the anti-schizophrenic effects of atypical antipsychotic drugs not only based on blockade of 5-HT2AR and of D2R in higher doses but also based on blocking the allosteric enhancement of D2R protomer signaling by 5-HT2AR protomer activation. This research opens a new understanding of the integration of DA and 5-HT signals released from DA and 5-HT nerve terminal networks. The biological principle of forming 5-HT and other heteroreceptor complexes in the brain also help understand the mechanism of action for especially the 5-HT hallucinogens, including putative positive effects of e.g., psilocybin and the indicated prosocial and anti-stress actions of MDMA (ecstasy). The GalR1-GalR2 heterodimer and the putative GalR1-GalR2-5-HT1 heteroreceptor complexes are targets for Galanin N-terminal fragment Gal (1-15), a major modulator of emotional networks in models of mental disease. GPCR-receptor tyrosine kinase (RTK) heteroreceptor complexes can operate through transactivation of FGFR1 via allosteric mechanisms and indirect interactions over GPCR intracellular pathways involving protein kinase Src which produces tyrosine phosphorylation of the RTK. The exciting discovery was made that several antidepressant drugs such as TCAs and SSRIs as well as the fast-acting antidepressant drug ketamine can directly bind to the TrkB receptor and provide a novel mechanism for their antidepressant actions. Understanding the role of astrocytes and their allosteric receptor-receptor interactions in modulating forebrain glutamate synapses with impact on dorsal raphe-forebrain serotonin neurons is also of high relevance for research on major depressive disorder.
Collapse
|
9
|
Petit-Pedrol M, Groc L. Regulation of membrane NMDA receptors by dynamics and protein interactions. J Cell Biol 2021; 220:211609. [PMID: 33337489 PMCID: PMC7754687 DOI: 10.1083/jcb.202006101] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/16/2022] Open
Abstract
Understanding neurotransmitter system crosstalk in the brain is a major challenge in neurobiology. Several intracellular and genomic cascades have been identified in this crosstalk. However, the discovery that neurotransmitter receptors are highly diffusive in the plasma membrane of neurons, where they form heterocomplexes with other proteins, has profoundly changed our view of neurotransmitter signaling. Here, we review new insights into neurotransmitter crosstalk at the plasma membrane. We focus on the membrane organization and interactome of the ionotropic glutamate N-methyl-D-aspartate receptor (NMDAR) that plays a central role in excitatory synaptic and network physiology and is involved in the etiology of several major neuropsychiatric disorders. The nanoscale organization and dynamics of NMDAR is a key regulatory process for glutamate synapse transmission, plasticity, and crosstalk with other neurotransmitter systems, such as the monoaminergic ones. The plasma membrane appears to be a prime regulatory compartment for spatial and temporal crosstalk between neurotransmitter systems in the healthy and diseased brain. Understanding the molecular mechanisms regulating membrane neurotransmitter receptor crosstalk will likely open research avenues for innovative therapeutical strategies.
Collapse
Affiliation(s)
- Mar Petit-Pedrol
- Université de Bordeaux, Centre National de la Recherche Scientifique, Interdisciplinary Institute for Neuroscience, Unité Mixte de Recherche 5297, Bordeaux, France
| | - Laurent Groc
- Université de Bordeaux, Centre National de la Recherche Scientifique, Interdisciplinary Institute for Neuroscience, Unité Mixte de Recherche 5297, Bordeaux, France
| |
Collapse
|
10
|
Misganaw D. Heteromerization of dopaminergic receptors in the brain: Pharmacological implications. Pharmacol Res 2021; 170:105600. [PMID: 33836279 DOI: 10.1016/j.phrs.2021.105600] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/17/2021] [Accepted: 04/02/2021] [Indexed: 12/15/2022]
Abstract
Dopamine exerts its physiological effects through two subtypes of receptors, i.e. the receptors of the D1 family (D1R and D5R) and the D2 family (D2R, D3R, and D4R), which differ in their pattern of distribution, affinity, and signaling. The D1-like subfamily (D1R and D5R) are coupled to Gαs/olf proteins to activate adenylyl cyclase whereas the D2-like receptors are coupled to Gαi/o subunits and suppress the activity of adenylyl cyclase. Dopamine receptors are capable of forming homodimers, heterodimers, and higher-order oligomeric complexes, resulting in a change in the individual protomers' recognition, signaling, and pharmacology. Heteromerization has the potential to modify the canonical pharmacological features of individual monomeric units such as ligand affinity, activation, signaling, and cellular trafficking through allosteric interactions, reviving the field and introducing a new pharmacological target. Since heteromers are expressed and formed in a tissue-specific manner, they could provide the framework to design selective and effective drug candidates, such as brain-penetrant heterobivalent drugs and interfering peptides, with limited side effects. Therefore, heteromerization could be a promising area of pharmacology research, as it could contribute to the development of novel pharmacological interventions for dopamine dysregulated brain disorders such as addiction, schizophrenia, cognition, Parkinson's disease, and other motor-related disorders. This review is articulated based on the three criteria established by the International Union of Basic and Clinical Pharmacology for GPCR heterodimers (IUPHAR): evidence of co-localization and physical interactions in native or primary tissue, presence of a new physiological and functional property than the individual protomers, and loss of interaction and functional fingerprints upon heterodimer disruption.
Collapse
Affiliation(s)
- Desye Misganaw
- Pharmacology and Toxicology Unit, Department of Pharmacy, College of Medicine and Health Science, Wollo University, P.O. Box 1145, Dessie, Ethiopia.
| |
Collapse
|
11
|
Menniti FS, Chappie TA, Schmidt CJ. PDE10A Inhibitors-Clinical Failure or Window Into Antipsychotic Drug Action? Front Neurosci 2021; 14:600178. [PMID: 33551724 PMCID: PMC7855852 DOI: 10.3389/fnins.2020.600178] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 12/21/2020] [Indexed: 01/21/2023] Open
Abstract
PDE10A, a phosphodiesterase that inactivates both cAMP and cGMP, is a unique signaling molecule in being highly and nearly exclusively expressed in striatal medium spiny neurons. These neurons dynamically integrate cortical information with dopamine-signaled value to mediate action selection among available behavioral options. Medium spiny neurons are components of either the direct or indirect striatal output pathways. Selective activation of indirect pathway medium spiny neurons by dopamine D2 receptor antagonists is putatively a key element in the mechanism of their antipsychotic efficacy. While PDE10A is expressed in all medium spiny neurons, studies in rodents indicated that PDE10A inhibition has behavioral effects in several key assays that phenocopy dopamine D2 receptor inhibition. This finding gave rise to the hypothesis that PDE10A inhibition also preferentially activates indirect pathway medium spiny neurons, a hypothesis that is consistent with electrophysiological, neurochemical, and molecular effects of PDE10A inhibitors. These data underwrote industry-wide efforts to investigate and develop PDE10A inhibitors as novel antipsychotics. Disappointingly, PDE10A inhibitors from 3 companies failed to evidence antipsychotic activity in patients with schizophrenia to the same extent as standard-of-care D2 antagonists. Given the notable similarities between PDE10A inhibitors and D2 antagonists, gaining an understanding of why only the latter class is antipsychotic affords a unique window into the basis for this therapeutic efficacy. With this in mind, we review the data on PDE10A inhibition as a step toward back-translating the limited antipsychotic efficacy of PDE10A inhibitors, hopefully to inform new efforts to develop better therapeutics to treat psychosis and schizophrenia.
Collapse
Affiliation(s)
- Frank S Menniti
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, United States
| | - Thomas A Chappie
- Internal Medicine Medicinal Chemistry, Pfizer Worldwide Research and Development, Cambridge, MA, United States
| | - Christopher J Schmidt
- Pfizer Innovation and Research Lab Unit, Pfizer Worldwide Research and Development, Cambridge, MA, United States
| |
Collapse
|
12
|
Gonçalves MCB, Glaser T, Oliveira SLBD, Ulrich H. Adenosinergic-Dopaminergic Signaling in Mood Disorders: A Mini-Review. J Caffeine Adenosine Res 2020. [DOI: 10.1089/caff.2020.0009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
| | - Talita Glaser
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | | | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, São Paulo, Brazil
| |
Collapse
|
13
|
Martel JC, Gatti McArthur S. Dopamine Receptor Subtypes, Physiology and Pharmacology: New Ligands and Concepts in Schizophrenia. Front Pharmacol 2020; 11:1003. [PMID: 32765257 PMCID: PMC7379027 DOI: 10.3389/fphar.2020.01003] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022] Open
Abstract
Dopamine receptors are widely distributed within the brain where they play critical modulator roles on motor functions, motivation and drive, as well as cognition. The identification of five genes coding for different dopamine receptor subtypes, pharmacologically grouped as D1- (D1 and D5) or D2-like (D2S, D2L, D3, and D4) has allowed the demonstration of differential receptor function in specific neurocircuits. Recent observation on dopamine receptor signaling point at dopamine-glutamate-NMDA neurobiology as the most relevant in schizophrenia and for the development of new therapies. Progress in the chemistry of D1- and D2-like receptor ligands (agonists, antagonists, and partial agonists) has provided more selective compounds possibly able to target the dopamine receptors homo and heterodimers and address different schizophrenia symptoms. Moreover, an extensive evaluation of the functional effect of these agents on dopamine receptor coupling and intracellular signaling highlights important differences that could also result in highly differentiated clinical pharmacology. The review summarizes the recent advances in the field, addressing the relevance of emerging new targets in schizophrenia in particular in relation to the dopamine - glutamate NMDA systems interactions.
Collapse
|
14
|
Multiple Adenosine-Dopamine (A2A-D2 Like) Heteroreceptor Complexes in the Brain and Their Role in Schizophrenia. Cells 2020; 9:cells9051077. [PMID: 32349279 PMCID: PMC7290895 DOI: 10.3390/cells9051077] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/20/2020] [Accepted: 04/20/2020] [Indexed: 12/21/2022] Open
Abstract
In the 1980s and 1990s, the concept was introduced that molecular integration in the Central Nervous System could develop through allosteric receptor–receptor interactions in heteroreceptor complexes presents in neurons. A number of adenosine–dopamine heteroreceptor complexes were identified that lead to the A2A-D2 heteromer hypothesis of schizophrenia. The hypothesis is based on strong antagonistic A2A-D2 receptor–receptor interactions and their presence in the ventral striato-pallidal GABA anti-reward neurons leading to reduction of positive symptoms. Other types of adenosine A2A heteroreceptor complexes are also discussed in relation to this disease, such as A2A-D3 and A2A-D4 heteroreceptor complexes as well as higher order A2A-D2-mGluR5 and A2A-D2-Sigma1R heteroreceptor complexes. The A2A receptor protomer can likely modulate the function of the D4 receptors of relevance for understanding cognitive dysfunction in schizophrenia. A2A-D2-mGluR5 complex is of interest since upon A2A/mGluR5 coactivation they appear to synergize in producing strong inhibition of the D2 receptor protomer. For understanding the future of the schizophrenia treatment, the vulnerability of the current A2A-D2like receptor complexes will be tested in animal models of schizophrenia. A2A-D2-Simag1R complexes hold the highest promise through Sigma1R enhancement of inhibition of D2R function. In line with this work, Lara proposed a highly relevant role of adenosine for neurobiology of schizophrenia.
Collapse
|
15
|
Nani JV, Yonamine CM, Castro Musial D, Dal Mas C, Mari JJ, Hayashi MAF. ACE activity in blood and brain axis in an animal model for schizophrenia: Effects of dopaminergic manipulation with antipsychotics and psychostimulants. World J Biol Psychiatry 2020; 21:53-63. [PMID: 30806143 DOI: 10.1080/15622975.2019.1583372] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Objectives: Angiotensin I-converting enzyme (ACE) was initially correlated with schizophrenia (SCZ) in studies showing a correlation of ACE increased enzyme activity with memory impairments. Possible role for ACE in SCZ was also suggested by ACE activity interaction with dopaminergic mechanisms to modulate abnormalities of sensorimotor gating. In addition, we have demonstrated higher ACE activity in blood of SCZ subjects, its implication in cognitive performance in SCZ and its power as a predictor for SCZ diagnosis.Methods: ACE activity was determined in the serum and in selected brain regions of an animal model presenting SCZ-like behaviour, before and after the treatment with typical and atypical antipsychotics, and also in the serum of animals receiving the psychostimulants amphetamine/lisdexamphetamine.Results: Dopaminergic manipulations with antipsychotics and psychostimulants influenced the ACE activity, but with no correlation with the animal blood pressure.Conclusions: The validity of measuring ACE activity in animal blood to predict activity in the CNS, as well as the lack of correlation between the activity and blood pressure, before and after the treatment with antipsychotics, were confirmed here. Correlations of the present findings with data from clinical studies also strengthen the value of this animal model for studying several aspects of SCZ.
Collapse
Affiliation(s)
- João V Nani
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.,Department of Psychiatry, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.,National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil
| | - Camila M Yonamine
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.,National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil
| | - Diego Castro Musial
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Caroline Dal Mas
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.,National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil
| | - Jair J Mari
- Department of Psychiatry, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Mirian A F Hayashi
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.,National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil
| |
Collapse
|
16
|
Bada Juarez JF, Muñoz-García JC, Inácio Dos Reis R, Henry A, McMillan D, Kriek M, Wood M, Vandenplas C, Sands Z, Castro L, Taylor R, Watts A. Detergent-free extraction of a functional low-expressing GPCR from a human cell line. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1862:183152. [PMID: 31843475 DOI: 10.1016/j.bbamem.2019.183152] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 11/21/2019] [Accepted: 12/05/2019] [Indexed: 01/02/2023]
Abstract
Dopamine receptors (DRs) are class A G-Protein Coupled Receptors (GPCRs) prevalent in the central nervous system (CNS). These receptors mediate physiological functions ranging from voluntary movement and reward recognition to hormonal regulation and hypertension. Drugs targeting dopaminergic neurotransmission have been employed to treat several neurological and psychiatric disorders, including Parkinson's disease, schizophrenia, Huntington's disease, attention deficit hyperactivity disorder (ADHD), and Tourette's syndrome. In vivo, incorporation of GPCRs into lipid membranes is known to be key to their biological function and, by inference, maintenance of their tertiary structure. A further significant challenge in the structural and biochemical characterization of human DRs is their low levels of expression in mammalian cells. Thus, the purification and enrichment of DRs whilst retaining their structural integrity and function is highly desirable for biophysical studies. A promising new approach is the use of styrene-maleic acid (SMA) copolymer to solubilize GPCRs directly in their native environment, to produce polymer-assembled Lipodisqs (LQs). We have developed a novel methodology to yield detergent-free D1-containing Lipodisqs directly from HEK293f cells expressing wild-type human dopamine receptor 1 (D1). We demonstrate that D1 in the Lipodisq retains activity comparable to that in the native environment and report, for the first time, the affinity constant for the interaction of the peptide neurotransmitter neurotensin (NT) with D1, in the native state.
Collapse
Affiliation(s)
| | - Juan C Muñoz-García
- Biochemistry Department, Oxford University, South Parks Road, Oxford OX1 3QU, UK; School of Pharmacy, University of East Anglia, Norwich NR4 7TJ, UK
| | | | | | | | - Marco Kriek
- UCB Celltech, 216 Bath Road, Slough SL1 3WE, UK
| | - Martyn Wood
- UCB BioPharma SPRL, Braine-l'Alleud, Belgium
| | | | - Zara Sands
- UCB Celltech, 216 Bath Road, Slough SL1 3WE, UK
| | - Luis Castro
- UCB Celltech, 216 Bath Road, Slough SL1 3WE, UK
| | | | - Anthony Watts
- Biochemistry Department, Oxford University, South Parks Road, Oxford OX1 3QU, UK.
| |
Collapse
|
17
|
Rodríguez B, Nani JV, Almeida PGC, Brietzke E, Lee RS, Hayashi MAF. Neuropeptides and oligopeptidases in schizophrenia. Neurosci Biobehav Rev 2019; 108:679-693. [PMID: 31794779 DOI: 10.1016/j.neubiorev.2019.11.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 11/14/2019] [Accepted: 11/27/2019] [Indexed: 12/30/2022]
Abstract
Schizophrenia (SCZ) is a complex psychiatric disorder with severe impact on patient's livelihood. In the last years, the importance of neuropeptides in SCZ and other CNS disorders has been recognized, mainly due to their ability to modulate the signaling of classical monoaminergic neurotransmitters as dopamine. In addition, a class of enzymes coined as oligopeptidases are able to cleave several of these neuropeptides, and their potential implication in SCZ was also demonstrated. Interestingly, these enzymes are able to play roles as modulators of neuropeptidergic systems, and they were also implicated in neurogenesis, neurite outgrowth, neuron migration, and therefore, in neurodevelopment and brain formation. Altered activity of oligopeptidases in SCZ was described only more recently, suggesting their possible utility as biomarkers for mental disorders diagnosis or treatment response. We provide here an updated and comprehensive review on neuropeptides and oligopeptidases involved in mental disorders, aiming to attract the attention of physicians to the potential of targeting this system for improving the therapy and for understanding the neurobiology underlying mental disorders as SCZ.
Collapse
Affiliation(s)
- Benjamín Rodríguez
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - João Victor Nani
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil; National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil
| | - Priscila G C Almeida
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Elisa Brietzke
- Department of Psychiatry, Queen's University School of Medicine, Kingston, ON, Canada
| | - Richard S Lee
- Department of Psychiatry, Johns Hopkins University, Baltimore, MD, USA
| | - Mirian A F Hayashi
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil; National Institute for Translational Medicine (INCT-TM, CNPq/FAPESP/CAPES), Ribeirão Preto, Brazil.
| |
Collapse
|
18
|
Plach M, Schäfer T, Borroto-Escuela DO, Weikert D, Gmeiner P, Fuxe K, Friedland K. Differential allosteric modulation within dopamine D 2R - neurotensin NTS1R and D 2R - serotonin 5-HT 2AR receptor complexes gives bias to intracellular calcium signalling. Sci Rep 2019; 9:16312. [PMID: 31704949 PMCID: PMC6841725 DOI: 10.1038/s41598-019-52540-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/15/2019] [Indexed: 12/11/2022] Open
Abstract
Proceeding investigations of G protein-coupled receptor (GPCR) heterocomplexes have demonstrated that the dopamine D2 receptor (D2R), one of the hub receptors in the physiology of schizophrenia, interacts with both the neurotensin NTS1 (NTS1R) and the serotonin 5-HT2A receptor (5-HT2AR) in cell lines and rodent brain tissue. In situ proximity ligation assay and BRET-based saturation experiments confirmed interacting receptor assemblies in HEK293T and neuronal HT22 cells. The NTS1R agonist NT(8-13) reduces the Gαq-mediated calcium signal in the NTS1R-D2R complex compared to the NTS1R monomer which could be reversed by D2R antagonists. The bivalent ligand CS148 (NTS1R-agonistic, D2R-antagonistic) increased the calcium response addressing the dimer, consistent with the effect of the monovalent ligands suggesting an allosteric D2R-mediated modulation. In contrast, the 5-HT2AR-D2R heteromer did not show a calcium-altering receptor-receptor interaction. Despite their common coupling-preference for Gαq, 5-HT2AR and NTS1R supposedly interact with D2R each in a unique mode. This remarkably diverse ligand-mediated signalling in two different D2R heteroreceptor complexes illustrates the complexity of receptor-receptor interactions and their potential of modifying cell responses to external stimuli. Therefore, GPCR heteromers may provide a very promising novel target for the therapy of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Michael Plach
- Department of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Thorsten Schäfer
- Department of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | | | - Dorothée Weikert
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Kristina Friedland
- Department of Chemistry and Pharmacy, Molecular and Clinical Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
- Pharmacology and Toxicology, Institute of Pharmacy and Biochemistry, Johannes-Gutenberg-Universität, Mainz, Germany.
| |
Collapse
|
19
|
Adenosine A2A receptor as potential therapeutic target in neuropsychiatric disorders. Pharmacol Res 2019; 147:104338. [DOI: 10.1016/j.phrs.2019.104338] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/26/2019] [Accepted: 07/01/2019] [Indexed: 01/20/2023]
|
20
|
León-Navarro DA, Albasanz JL, Martín M. Functional Cross-Talk between Adenosine and Metabotropic Glutamate Receptors. Curr Neuropharmacol 2019; 17:422-437. [PMID: 29663888 PMCID: PMC6520591 DOI: 10.2174/1570159x16666180416093717] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/19/2018] [Accepted: 04/13/2018] [Indexed: 12/14/2022] Open
Abstract
Abstract: G-protein coupled receptors are transmembrane proteins widely expressed in cells and their transduction pathways are mediated by controlling second messenger levels through different G-protein interactions. Many of these receptors have been described as involved in the physiopathology of neurodegenerative diseases and even considered as potential targets for the design of novel therapeutic strategies. Endogenous and synthetic allosteric and orthosteric selective ligands are able to modulate GPCRs at both gene and protein expression levels and can also modify their physiological function. GPCRs that coexist in the same cells can homo- and heteromerize, therefore, modulating their function. Adenosine receptors are GPCRs which stimulate or inhibit adenylyl cyclase activity through Gi/Gs protein and are involved in the control of neurotransmitter release as glutamate. In turn, metabotropic glutamate receptors are also GPCRs which inhibit adenylyl cyclase or stimulate phospholipase C activities through Gi or Gq proteins, respectively. In recent years, evidence of crosstalk mechanisms be-tween different GPCRs have been described. The aim of the present review was to summarize the described mechanisms of interaction and crosstalking between adenosine and metabotropic glutamate receptors, mainly of group I, in both in vitro and in vivo systems, and their possible use for the design of novel ligands for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- David Agustín León-Navarro
- Departamento de Quimica Inorganica, Organica y Bioquimica. CRIB, Universidad de Castilla-La Mancha, Spain.,Facultad de Ciencias y Tecnologías Químicas, Avenida Camilo José Cela, 10, 13071 Ciudad Real, Spain
| | - José Luis Albasanz
- Departamento de Quimica Inorganica, Organica y Bioquimica. CRIB, Universidad de Castilla-La Mancha, Spain.,Facultad de Ciencias y Tecnologías Químicas, Avenida Camilo José Cela, 10, 13071 Ciudad Real, Spain.,Facultad de Medicina de Ciudad Real, Camino Moledores s/n. 13071 Ciudad Real, Spain
| | - Mairena Martín
- Departamento de Quimica Inorganica, Organica y Bioquimica. CRIB, Universidad de Castilla-La Mancha, Spain.,Facultad de Ciencias y Tecnologías Químicas, Avenida Camilo José Cela, 10, 13071 Ciudad Real, Spain.,Facultad de Medicina de Ciudad Real, Camino Moledores s/n. 13071 Ciudad Real, Spain
| |
Collapse
|
21
|
Fuxe K, Borroto-Escuela DO. Understanding receptor heteromerization and its allosteric integration of signals. Neuropharmacology 2019; 152:1-3. [PMID: 31054939 DOI: 10.1016/j.neuropharm.2019.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Dasiel O Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Biomolecular Science, Section of Physiology, University of Urbino, Campus Scientifico Enrico Mattei, via Ca' le Suore 2, I-61029, Urbino, Italy; Observatorio Cubano de Neurociencias, Grupo Bohío-Estudio, Zayas 50, 62100, Yaguajay, Cuba.
| |
Collapse
|
22
|
Effects of a methanol extract of Ficus platyphylla stem bark on a two-way active avoidance task and on body core temperature. Behav Brain Res 2019; 367:215-220. [DOI: 10.1016/j.bbr.2019.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/04/2019] [Accepted: 04/06/2019] [Indexed: 12/16/2022]
|
23
|
Al-Shar'i NA, Al-Balas QA. Molecular Dynamics Simulations of Adenosine Receptors: Advances, Applications and Trends. Curr Pharm Des 2019; 25:783-816. [DOI: 10.2174/1381612825666190304123414] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 02/26/2019] [Indexed: 01/09/2023]
Abstract
:
Adenosine receptors (ARs) are transmembrane proteins that belong to the G protein-coupled receptors
(GPCRs) superfamily and mediate the biological functions of adenosine. To date, four AR subtypes are known,
namely A1, A2A, A2B and A3 that exhibit different signaling pathways, tissue localization, and mechanisms of
activation. Moreover, the widespread ARs and their implication in numerous physiological and pathophysiological
conditions had made them pivotal therapeutic targets for developing clinically effective agents.
:
The crystallographic success in identifying the 3D crystal structures of A2A and A1 ARs has dramatically enriched
our understanding of their structural and functional properties such as ligand binding and signal transduction.
This, in turn, has provided a structural basis for a larger contribution of computational methods, particularly molecular
dynamics (MD) simulations, toward further investigation of their molecular properties and designing
bioactive ligands with therapeutic potential. MD simulation has been proved to be an invaluable tool in investigating
ARs and providing answers to some critical questions. For example, MD has been applied in studying ARs
in terms of ligand-receptor interactions, molecular recognition, allosteric modulations, dimerization, and mechanisms
of activation, collectively aiding in the design of subtype selective ligands.
:
In this review, we focused on the advances and different applications of MD simulations utilized to study the
structural and functional aspects of ARs that can foster the structure-based design of drug candidates. In addition,
relevant literature was briefly discussed which establishes a starting point for future advances in the field of drug
discovery to this pivotal group of drug targets.
Collapse
Affiliation(s)
- Nizar A. Al-Shar'i
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| | - Qosay A. Al-Balas
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| |
Collapse
|
24
|
Monte GG, Nani JV, de Almeida Campos MR, Dal Mas C, Marins LAN, Martins LG, Tasic L, Mori MA, Hayashi MAF. Impact of nuclear distribution element genes in the typical and atypical antipsychotics effects on nematode Caenorhabditis elegans: Putative animal model for studying the pathways correlated to schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:19-30. [PMID: 30578843 DOI: 10.1016/j.pnpbp.2018.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/17/2018] [Accepted: 12/19/2018] [Indexed: 12/18/2022]
Abstract
The nuclear distribution element genes are conserved from fungus to humans. The nematode Caenorhabditis elegans expresses two isoforms of nuclear distribution element genes, namely nud-1 and nud-2. While nud-1 was functionally demonstrated to be the worm nudC ortholog, bioinformatic analysis revealed that the nud-2 gene encodes the worm ortholog of the mammalian NDE1 (Nuclear Distribution Element 1 or NudE) and NDEL1 (NDE-Like 1 or NudEL) genes, which share overlapping roles in brain development in mammals and also mediate the axon guidance in mammalian and C. elegans neurons. A significantly higher NDEL1 enzyme activity was shown in treatment non-resistant compared to treatment resistant SCZ patients, who essentially present response to the therapy with atypical clozapine but not with typical antipsychotics. Using C. elegans as a model, we tested the consequence of nud genes suppression in the effects of typical and atypical antipsychotics. To assess the role of nud genes and antipsychotic drugs over C. elegans behavior, we measured body bend frequency, egg laying and pharyngeal pumping, which traits are controlled by specific neurons and neurotransmitters known to be involved in SCZ, as dopamine and serotonin. Evaluation of metabolic and behavioral response to the pharmacotherapy with these antipsychotics demonstrates an important unbalance in serotonin pathway in both nud-1 and nud-2 knockout worms, with more significant effects for nud-2 knockout. The present data also show an interesting trend of mutant knockout worm strains to present a metabolic profile closer to that observed for the wild-type animals after the treatment with the typical antipsychotic haloperidol, but which was not observed for the treatment with the atypical antipsychotic clozapine. Paradoxically, behavioral assays showed more evident effects for clozapine than for haloperidol, which is in line with previous studies with rodent animal models and clinical evaluations with SCZ patients. In addition, the validity and reliability of using this experimental animal model to further explore the convergence between the dopamine/serotonin pathways and neurodevelopmental processes was demonstrated here, and the potential usefulness of this model for evaluating the metabolic consequences of treatments with antipsychotics is also suggested.
Collapse
Affiliation(s)
- Gabriela Guilherme Monte
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP) São Paulo, Brazil
| | - João V Nani
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP) São Paulo, Brazil
| | | | - Caroline Dal Mas
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP) São Paulo, Brazil
| | - Lucas Augusto Negri Marins
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP) São Paulo, Brazil
| | - Lucas Gelain Martins
- Chemical Biology Laboratory, Department of Organic Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Ljubica Tasic
- Chemical Biology Laboratory, Department of Organic Chemistry, Institute of Chemistry, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Marcelo A Mori
- Departament of Biochemistry and Tissue Biology, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | - Mirian A F Hayashi
- Department of Pharmacology, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP) São Paulo, Brazil.
| |
Collapse
|
25
|
Schwitzer T, Schwan R, Angioi-Duprez K, Lalanne L, Giersch A, Laprevote V. Cannabis use and human retina: The path for the study of brain synaptic transmission dysfunctions in cannabis users. Neurosci Biobehav Rev 2019; 106:11-22. [PMID: 30773228 DOI: 10.1016/j.neubiorev.2018.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 11/08/2018] [Accepted: 12/02/2018] [Indexed: 01/01/2023]
Abstract
Owing to the difficulty of obtaining direct access to the functioning brain, new approaches are needed for the indirect exploration of brain disorders in neuroscience research. Due to its embryonic origin, the retina is part of the central nervous system and is well suited to the investigation of neurological functions in psychiatric and addictive disorders. In this review, we focus on cannabis use, which is a crucial public health challenge, since cannabis is one of the most widely used addictive drugs in industrialized countries. We first explain why studying retinal function is relevant when exploring the effects of cannabis use on brain function. Next, we describe both the retinal electrophysiological measurements and retinal dysfunctions observed after acute and regular cannabis use. We then discuss how these retinal dysfunctions may inform brain synaptic transmission abnormalities. Finally, we present various directions for future research on the neurotoxic effects of cannabis use.
Collapse
Affiliation(s)
- Thomas Schwitzer
- Pôle Hospitalo-Universitaire de Psychiatrie d'Adultes du Grand Nancy, Centre Psychothérapique de Nancy, Laxou, France; INSERM U1114, Fédération de Médecine Translationnelle de Strasbourg, Département de Psychiatrie, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France.
| | - Raymund Schwan
- Pôle Hospitalo-Universitaire de Psychiatrie d'Adultes du Grand Nancy, Centre Psychothérapique de Nancy, Laxou, France; INSERM U1114, Fédération de Médecine Translationnelle de Strasbourg, Département de Psychiatrie, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France; Maison des Addictions, CHRU Nancy, Nancy, France
| | | | - Laurence Lalanne
- INSERM U1114, Fédération de Médecine Translationnelle de Strasbourg, Département de Psychiatrie, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France; Pôle de Psychiatrie Santé Mentale et Addictologie, Fédération de Médecine Translationnelle de Strasbourg, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France
| | - Anne Giersch
- INSERM U1114, Fédération de Médecine Translationnelle de Strasbourg, Département de Psychiatrie, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France
| | - Vincent Laprevote
- Pôle Hospitalo-Universitaire de Psychiatrie d'Adultes du Grand Nancy, Centre Psychothérapique de Nancy, Laxou, France; INSERM U1114, Fédération de Médecine Translationnelle de Strasbourg, Département de Psychiatrie, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France
| |
Collapse
|
26
|
Lewis MH, Primiani CT, Muehlmann AM. Targeting Dopamine D 2, Adenosine A 2A, and Glutamate mGlu 5 Receptors to Reduce Repetitive Behaviors in Deer Mice. J Pharmacol Exp Ther 2019; 369:88-97. [PMID: 30745415 DOI: 10.1124/jpet.118.256081] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 02/07/2019] [Indexed: 12/19/2022] Open
Abstract
Repetitive behaviors are seemingly purposeless patterns of behavior that vary little in form and are characteristic of many neurodevelopmental, psychiatric, and neurologic disorders. Our work has identified an association between hypofunctioning of the indirect basal ganglia pathway and the expression of repetitive behavior in the deer mouse model. In this study, we targeted indirect pathway cells of the striatum with single drugs and drug combinations that bind to dopamine D2, adenosine A2A, and glutamate mGlu5 receptors. These receptors function both individually and as receptor heteromers. We found that only the triple drug cocktail (L-741,626+CGS21680+CDPPB) that was designed to increase striatal indirect basal ganglia pathway cell function reduced repetitive behavior in adult male deer mice. No single drug or double drug combinations were effective at selectively reducing repetitive behavior. We found this triple drug cocktail reduced repetitive behavior in both short-acting and long-acting formulations and was effective throughout 7 days of daily administration. Conversely, another triple drug cocktail (quinpirole+SCH58261+MTEP) that was designed to further reduce striatal indirect basal ganglia pathway cell function caused a significant increase in repetitive behavior. Significant and behaviorally selective effects on repetitive behavior were only achieved with the triple drug cocktails that included doses of L-741,626 and quinpirole that have off-target effects (e.g., dopamine D3 receptors). These data further a role for decreased indirect basal ganglia pathway activation in repetitive behavior and suggest that targeting these receptors and/or heteromeric complexes on the indirect pathway neurons of the striatum may offer pharmacotherapeutic benefit for individuals with repetitive behavior disorders.
Collapse
Affiliation(s)
- Mark H Lewis
- Department of Psychiatry, University of Florida, Gainesville, Florida
| | | | - Amber M Muehlmann
- Department of Psychiatry, University of Florida, Gainesville, Florida
| |
Collapse
|
27
|
Derouiche L, Massotte D. G protein-coupled receptor heteromers are key players in substance use disorder. Neurosci Biobehav Rev 2018; 106:73-90. [PMID: 30278192 DOI: 10.1016/j.neubiorev.2018.09.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 09/25/2018] [Accepted: 09/26/2018] [Indexed: 12/19/2022]
Abstract
G protein-coupled receptors (GPCR) represent the largest family of membrane proteins in the human genome. Physical association between two different GPCRs is linked to functional interactions which generates a novel entity, called heteromer, with specific ligand binding and signaling properties. Heteromerization is increasingly recognized to take place in the mesocorticolimbic pathway and to contribute to various aspects related to substance use disorder. This review focuses on heteromers identified in brain areas relevant to drug addiction. We report changes at the molecular and cellular levels that establish specific functional impact and highlight behavioral outcome in preclinical models. Finally, we briefly discuss selective targeting of native heteromers as an innovative therapeutic option.
Collapse
Affiliation(s)
- Lyes Derouiche
- Institut des Neurosciences Cellulaires et Integratives, UPR 3212, 5 rue Blaise Pascal, F-67000 Strasbourg, France
| | - Dominique Massotte
- Institut des Neurosciences Cellulaires et Integratives, UPR 3212, 5 rue Blaise Pascal, F-67000 Strasbourg, France.
| |
Collapse
|
28
|
Abstract
Initially G protein-coupled receptors, GPCRs, were thought to act as monomers, but recently strong evidence has been gathered indicating that they are capable of forming homo- and heterodimers or higher order oligomeric complexes, and that the dimerization phenomenon can modulate the pharmacological response and function of these receptors. In this chapter we point to the great potential of alternative therapeutic approach targeted at GPCR dimers, which is especially important in the field of neuropsychopharmacology. We also included a brief description of methods used for studying the phenomenon of GPCR oligomerization, with particular attention paid to the proximity ligation assay, PLA, the procedure which allows the study of interactions between receptors not only in vitro but also in vivo, with good anatomical resolution, what is especially important in the studies of various GPCRs involved in central neurotransmission.
Collapse
|
29
|
Borroto-Escuela DO, Rodriguez D, Romero-Fernandez W, Kapla J, Jaiteh M, Ranganathan A, Lazarova T, Fuxe K, Carlsson J. Mapping the Interface of a GPCR Dimer: A Structural Model of the A 2A Adenosine and D 2 Dopamine Receptor Heteromer. Front Pharmacol 2018; 9:829. [PMID: 30214407 PMCID: PMC6125358 DOI: 10.3389/fphar.2018.00829] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 07/10/2018] [Indexed: 12/17/2022] Open
Abstract
The A2A adenosine (A2AR) and D2 dopamine (D2R) receptors form oligomers in the cell membrane and allosteric interactions across the A2AR–D2R heteromer represent a target for development of drugs against central nervous system disorders. However, understanding of the molecular determinants of A2AR–D2R heteromerization and the allosteric antagonistic interactions between the receptor protomers is still limited. In this work, a structural model of the A2AR–D2R heterodimer was generated using a combined experimental and computational approach. Regions involved in the heteromer interface were modeled based on the effects of peptides derived from the transmembrane (TM) helices on A2AR–D2R receptor–receptor interactions in bioluminescence resonance energy transfer (BRET) and proximity ligation assays. Peptides corresponding to TM-IV and TM-V of the A2AR blocked heterodimer interactions and disrupted the allosteric effect of A2AR activation on D2R agonist binding. Protein–protein docking was used to construct a model of the A2AR–D2R heterodimer with a TM-IV/V interface, which was refined using molecular dynamics simulations. Mutations in the predicted interface reduced A2AR–D2R interactions in BRET experiments and altered the allosteric modulation. The heterodimer model provided insights into the structural basis of allosteric modulation and the technique developed to characterize the A2AR–D2R interface can be extended to study the many other G protein-coupled receptors that engage in heteroreceptor complexes.
Collapse
Affiliation(s)
| | - David Rodriguez
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Wilber Romero-Fernandez
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Jon Kapla
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Mariama Jaiteh
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Anirudh Ranganathan
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Tzvetana Lazarova
- Department of Biochemistry and Molecular Biology, Institute of Neuroscience, Faculty of Medicine, Autonomous University of Barcelona, Barcelona, Spain
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Jens Carlsson
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
30
|
Borroto-Escuela DO, Perez De La Mora M, Manger P, Narváez M, Beggiato S, Crespo-Ramírez M, Navarro G, Wydra K, Díaz-Cabiale Z, Rivera A, Ferraro L, Tanganelli S, Filip M, Franco R, Fuxe K. Brain Dopamine Transmission in Health and Parkinson's Disease: Modulation of Synaptic Transmission and Plasticity Through Volume Transmission and Dopamine Heteroreceptors. Front Synaptic Neurosci 2018; 10:20. [PMID: 30042672 PMCID: PMC6048293 DOI: 10.3389/fnsyn.2018.00020] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 06/19/2018] [Indexed: 01/04/2023] Open
Abstract
This perspective article provides observations supporting the view that nigro-striatal dopamine neurons and meso-limbic dopamine neurons mainly communicate through short distance volume transmission in the um range with dopamine diffusing into extrasynaptic and synaptic regions of glutamate and GABA synapses. Based on this communication it is discussed how volume transmission modulates synaptic glutamate transmission onto the D1R modulated direct and D2R modulated indirect GABA pathways of the dorsal striatum. Each nigro-striatal dopamine neuron was first calculated to form large numbers of neostriatal DA nerve terminals and then found to give rise to dense axonal arborizations spread over the neostriatum, from which dopamine is released. These neurons can through DA volume transmission directly influence not only the striatal GABA projection neurons but all the striatal cell types in parallel. It includes the GABA nerve cells forming the island-/striosome GABA pathway to the nigral dopamine cells, the striatal cholinergic interneurons and the striatal GABA interneurons. The dopamine modulation of the different striatal nerve cell types involves the five dopamine receptor subtypes, D1R to D5R receptors, and their formation of multiple extrasynaptic and synaptic dopamine homo and heteroreceptor complexes. These features of the nigro-striatal dopamine neuron to modulate in parallel the activity of practically all the striatal nerve cell types in the dorsal striatum, through the dopamine receptor complexes allows us to understand its unique and crucial fine-tuning of movements, which is lost in Parkinson's disease. Integration of striatal dopamine signals with other transmitter systems in the striatum mainly takes place via the receptor-receptor interactions in dopamine heteroreceptor complexes. Such molecular events also participate in the integration of volume transmission and synaptic transmission. Dopamine modulation of the glutamate synapses on the dorsal striato-pallidal GABA pathway involves D2R heteroreceptor complexes such as D2R-NMDAR, A2AR-D2R, and NTSR1-D2R heteroreceptor complexes. The dopamine modulation of glutamate synapses on the striato-entopeduncular/nigral pathway takes place mainly via D1R heteroreceptor complexes such as D1R-NMDAR, A2R-D1R, and D1R-D3R heteroreceptor complexes. Dopamine modulation of the island/striosome compartment of the dorsal striatum projecting to the nigral dopamine cells involve D4R-MOR heteroreceptor complexes. All these receptor-receptor interactions have relevance for Parkinson's disease and its treatment.
Collapse
Affiliation(s)
- Dasiel O. Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Section of Physiology, Department of Biomolecular Science, University of Urbino, Urbino, Italy
- Observatorio Cubano de Neurociencias, Grupo Bohío-Estudio, Yaguajay, Cuba
| | - Miguel Perez De La Mora
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Paul Manger
- Faculty of Health Sciences, School of Anatomical Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Manuel Narváez
- Facultad de Medicina, Instituto de Investigación Biomédica de Málaga, Málaga, Spain
| | - Sarah Beggiato
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Minerva Crespo-Ramírez
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gemma Navarro
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biomedicine, University of Barcelona, Barcelona, Spain
| | - Karolina Wydra
- Laboratory of Drug Addiction Pharmacology, Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Zaida Díaz-Cabiale
- Facultad de Medicina, Instituto de Investigación Biomédica de Málaga, Málaga, Spain
| | - Alicia Rivera
- Department of Cell Biology, Faculty of Sciences, University of Málaga, Málaga, Spain
| | - Luca Ferraro
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Sergio Tanganelli
- Department of Life Sciences and Biotechnology (SVEB), University of Ferrara, Ferrara, Italy
| | - Małgorzata Filip
- Laboratory of Drug Addiction Pharmacology, Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Rafael Franco
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biomedicine, University of Barcelona, Barcelona, Spain
- CiberNed: Centro de Investigación en Red Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid, Spain
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
31
|
Aringhieri S, Carli M, Kolachalam S, Verdesca V, Cini E, Rossi M, McCormick PJ, Corsini GU, Maggio R, Scarselli M. Molecular targets of atypical antipsychotics: From mechanism of action to clinical differences. Pharmacol Ther 2018; 192:20-41. [PMID: 29953902 DOI: 10.1016/j.pharmthera.2018.06.012] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The introduction of atypical antipsychotics (AAPs) since the discovery of its prototypical drug clozapine has been a revolutionary pharmacological step for treating psychotic patients as these allow a significant recovery not only in terms of hospitalization and reduction in symptoms severity, but also in terms of safety, socialization and better rehabilitation in the society. Regarding the mechanism of action, AAPs are weak D2 receptor antagonists and they act beyond D2 antagonism, involving other receptor targets which regulate dopamine and other neurotransmitters. Consequently, AAPs present a significant reduction of deleterious side effects like parkinsonism, hyperprolactinemia, apathy and anhedonia, which are all linked to the strong blockade of D2 receptors. This review revisits previous and current findings within the class of AAPs and highlights the differences in terms of receptor properties and clinical activities among them. Furthermore, we propose a continuum spectrum of "atypia" that begins with risperidone (the least atypical) to clozapine (the most atypical), while all the other AAPs fall within the extremes of this spectrum. Clozapine is still considered the gold standard in refractory schizophrenia and in psychoses present in Parkinson's disease, though it has been associated with adverse effects like agranulocytosis (0.7%) and weight gain, pushing the scientific community to find new drugs as effective as clozapine, but devoid of its side effects. To achieve this, it is therefore imperative to characterize and compare in depth the very complex molecular profile of AAPs. We also introduce relatively new concepts like biased agonism, receptor dimerization and neurogenesis to identify better the old and new hallmarks of "atypia". Finally, a detailed confrontation of clinical differences among the AAPs is presented, especially in relation to their molecular targets, and new means like therapeutic drug monitoring are also proposed to improve the effectiveness of AAPs in clinical practice.
Collapse
Affiliation(s)
- Stefano Aringhieri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Marco Carli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Shivakumar Kolachalam
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Valeria Verdesca
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Enrico Cini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Mario Rossi
- Institute of Molecular Cell and Systems Biology, University of Glasgow, UK
| | - Peter J McCormick
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK
| | - Giovanni U Corsini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Roberto Maggio
- Biotechnological and Applied Clinical Sciences Department, University of L'Aquila, Italy
| | - Marco Scarselli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy.
| |
Collapse
|
32
|
Kostrzewa RM, Wydra K, Filip M, Crawford CA, McDougall SA, Brown RW, Borroto-Escuela DO, Fuxe K, Gainetdinov RR. Dopamine D 2 Receptor Supersensitivity as a Spectrum of Neurotoxicity and Status in Psychiatric Disorders. J Pharmacol Exp Ther 2018; 366:519-526. [PMID: 29921706 DOI: 10.1124/jpet.118.247981] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/23/2018] [Indexed: 12/13/2022] Open
Abstract
Abnormality of dopamine D2 receptor (D2R) function, often observed as D2R supersensitivity (D2RSS), is a commonality of schizophrenia and related psychiatric disorders in humans. Moreover, virtually all psychotherapeutic agents for schizophrenia target D2R in brain. Permanent D2RSS as a feature of a new animal model of schizophrenia was first reported in 1991, and then behaviorally and biochemically characterized over the next 15-20 years. In this model of schizophrenia characterized by production of D2RSS in ontogeny, there are demonstrated alterations of signaling processes, as well as functional links between the biologic template of the animal model and ability of pharmacotherapeutics to modulate or reverse biologic and behavioral modalities toward normality. Another such animal model, featuring knockout of trace amine-associated receptor 1 (TAAR1), demonstrates D2RSS with an increase in the proportion of D2R in the high-affinity state. Currently, TAAR1 agonists are being explored as a therapeutic option for schizophrenia. There is likewise an overlay of D2RSS with substance use disorder. The aspect of adenosine A2A-D2 heteroreceptor complexes in substance use disorder is highlighted, and the association of adenosine A2A receptor antagonists in discriminative and rewarding effects of psychostimulants is outlined. In summary, these new animal models of schizophrenia have face, construct, and predictive validity, and distinct advantages over earlier models. While the review summarizes elements of D2RSS in schizophrenia per se, and its interplay with substance use disorder, a major focus is on presumed new molecular targets attending D2RSS in schizophrenia and related clinical entities.
Collapse
Affiliation(s)
- Richard M Kostrzewa
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee (R.M.K., R.W.B.); Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Krakow, Poland (K.W., M.F.); Department of Psychology, California State University, San Bernardino, California (C.A.C., S.A.M.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (D.O.B.-E., K.F.); Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); and Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia (R.R.G.)
| | - Karolina Wydra
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee (R.M.K., R.W.B.); Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Krakow, Poland (K.W., M.F.); Department of Psychology, California State University, San Bernardino, California (C.A.C., S.A.M.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (D.O.B.-E., K.F.); Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); and Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia (R.R.G.)
| | - Malgorzata Filip
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee (R.M.K., R.W.B.); Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Krakow, Poland (K.W., M.F.); Department of Psychology, California State University, San Bernardino, California (C.A.C., S.A.M.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (D.O.B.-E., K.F.); Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); and Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia (R.R.G.)
| | - Cynthia A Crawford
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee (R.M.K., R.W.B.); Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Krakow, Poland (K.W., M.F.); Department of Psychology, California State University, San Bernardino, California (C.A.C., S.A.M.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (D.O.B.-E., K.F.); Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); and Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia (R.R.G.)
| | - Sanders A McDougall
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee (R.M.K., R.W.B.); Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Krakow, Poland (K.W., M.F.); Department of Psychology, California State University, San Bernardino, California (C.A.C., S.A.M.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (D.O.B.-E., K.F.); Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); and Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia (R.R.G.)
| | - Russell W Brown
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee (R.M.K., R.W.B.); Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Krakow, Poland (K.W., M.F.); Department of Psychology, California State University, San Bernardino, California (C.A.C., S.A.M.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (D.O.B.-E., K.F.); Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); and Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia (R.R.G.)
| | - Dasiel O Borroto-Escuela
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee (R.M.K., R.W.B.); Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Krakow, Poland (K.W., M.F.); Department of Psychology, California State University, San Bernardino, California (C.A.C., S.A.M.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (D.O.B.-E., K.F.); Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); and Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia (R.R.G.)
| | - Kjell Fuxe
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee (R.M.K., R.W.B.); Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Krakow, Poland (K.W., M.F.); Department of Psychology, California State University, San Bernardino, California (C.A.C., S.A.M.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (D.O.B.-E., K.F.); Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); and Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia (R.R.G.)
| | - Raul R Gainetdinov
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee (R.M.K., R.W.B.); Institute of Pharmacology, Polish Academy of Sciences, Department of Drug Addiction Pharmacology, Krakow, Poland (K.W., M.F.); Department of Psychology, California State University, San Bernardino, California (C.A.C., S.A.M.); Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden (D.O.B.-E., K.F.); Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia (R.R.G.); and Skolkovo Institute of Science and Technology, Skolkovo, Moscow, Russia (R.R.G.)
| |
Collapse
|
33
|
Borroto-Escuela DO, Tarakanov AO, Brito I, Fuxe K. Glutamate heteroreceptor complexes in the brain. Pharmacol Rep 2018; 70:936-950. [PMID: 32002960 DOI: 10.1016/j.pharep.2018.04.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 03/26/2018] [Accepted: 04/09/2018] [Indexed: 10/17/2022]
Abstract
The existence of mGluR, NMDAR, AMPAR and putative KAR heteroreceptor complexes in synaptic and extrasynaptic regions of brain glutamate synapses represents a major integrative mechanism. Our aim in the current article is to analyze if the formation of the different types glutamate hetereceptor complexes involves the contribution of triplet amino acid homologies (protriplets) in a postulated receptor interface based on the triplet puzzle theory. Seven main sets (lists) of receptor pairs in databases were used containing various sets (lists) of human receptor heteromers and nonheteromers obtained from the available scientific publications including the publically available GPCR-hetnet database. Brain mGluR1-mGluR5 and mGluR2-mGluR4 isoreceptor complexes were demonstrated with a predominant extrasynaptic localization at a post- and prejunctional localization. The existence of putative mGluR4-mGluR7 heteroreceptor complexes in the basal ganglia is proposed. Metabotropic glutamate receptor subtypes also participated in the formation of a large number of heteroreceptor complexes like mGluR1-A1R, mGluR5-A2AR, mGluR5-D2R and D2R-A2AR-mGluR5, located in relation to glutamate synapses, especially in the basal ganglia. A putative mGluR1-GABAB1/2 heterocomplex may also exist. NMDAR heteroreceptor complexes were also demonstrated as a fundamental integrative mechanism in the glutamate synapse and its extrasynaptic membranes. It represented fundamental work on inter alia NMDAR-mGluR5, NMDAR-D1R and NMDAR-D2R heteroreceptor complexes involving both antagonistic and facilitatory allosteric receptor-receptor interactions. As to AMPA receptors, a heterocomplex was found for the interaction between IFNgR1 and the AMPAR mediated via the subunit GluA1 which may be of relevance for neuroinflammation. AMPAR-D2R heteroreceptor complexes were also demonstrated. Besides glutamate heteroreceptor complexes and their allosteric receptor-receptor interactions, a significant mechanism for the functional crosstalk can also be phosphorylation and/or reorganization of adapter proteins with dynamic binding to the two receptors modulating the allosteric receptor mechanism.
Collapse
Affiliation(s)
- Dasiel O Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Biomolecular Science, Section of Physiology, University of Urbino, Campus Scientifico Enrico Mattei, Urbino, Italy.,Grupo Bohío-Estudio, Observatorio Cubano de Neurociencias, Yaguajay, Cuba
| | - Alexander O Tarakanov
- St. Petersburg Institute for Informatics and Automation, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Ismel Brito
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Grupo Bohío-Estudio, Observatorio Cubano de Neurociencias, Yaguajay, Cuba
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
34
|
Real JI, Simões AP, Cunha RA, Ferreira SG, Rial D. Adenosine A 2A receptors modulate the dopamine D 2 receptor-mediated inhibition of synaptic transmission in the mouse prefrontal cortex. Eur J Neurosci 2018; 47:1127-1134. [PMID: 29570875 DOI: 10.1111/ejn.13912] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/13/2018] [Accepted: 03/13/2018] [Indexed: 01/20/2023]
Abstract
Prefrontal cortex (PFC) circuits are modulated by dopamine acting on D1 - and D2 -like receptors, which are pharmacologically exploited to manage neuropsychiatric conditions. Adenosine A2A receptors (A2A R) also control PFC-related responses and A2A R antagonists are potential anti-psychotic drugs. As tight antagonistic A2A R-D2 R and synergistic A2A R-D1 R interactions occur in other brain regions, we now investigated the crosstalk between A2A R and D1 /D2 R controlling synaptic transmission between layers II/III and V in mouse PFC coronal slices. Dopamine decreased synaptic transmission, a presynaptic effect based on the parallel increase in paired-pulse responses. Dopamine inhibition was prevented by the D2 R-like antagonist sulpiride but not by the D1 R antagonist SCH23390 and was mimicked by the D2 R agonist sumanirole, but not by the agonists of either D4 R (A-412997) or D3 R (PD128907). Dopamine inhibition was prevented by the A2A R antagonist, SCH58261, and attenuated in A2A R knockout mice. Accordingly, triple-labelling immunocytochemistry experiments revealed the co-localization of A2A R and D2 R immunoreactivity in glutamatergic (vGluT1-positive) nerve terminals of the PFC. This reported positive A2A R-D2 R interaction controlling PFC synaptic transmission provides a mechanistic justification for the anti-psychotic potential of A2A R antagonists.
Collapse
Affiliation(s)
- Joana I Real
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Polo I, Rua Larga, 3004-504, Coimbra, Portugal
| | - Ana Patrícia Simões
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Polo I, Rua Larga, 3004-504, Coimbra, Portugal
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Polo I, Rua Larga, 3004-504, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Samira G Ferreira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Polo I, Rua Larga, 3004-504, Coimbra, Portugal
| | - Daniel Rial
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Polo I, Rua Larga, 3004-504, Coimbra, Portugal
| |
Collapse
|
35
|
Szlachta M, Kuśmider M, Pabian P, Solich J, Kolasa M, Żurawek D, Dziedzicka-Wasylewska M, Faron-Górecka A. Repeated Clozapine Increases the Level of Serotonin 5-HT 1AR Heterodimerization with 5-HT 2A or Dopamine D 2 Receptors in the Mouse Cortex. Front Mol Neurosci 2018; 11:40. [PMID: 29497362 PMCID: PMC5818438 DOI: 10.3389/fnmol.2018.00040] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 01/29/2018] [Indexed: 12/20/2022] Open
Abstract
G-protein–coupled receptor (GPCR) heterodimers are new targets for the treatment of schizophrenia. Dopamine D2 receptors and serotonin 5-HT1A and 5-HT2A receptors play an important role in neurotransmission and have been implicated in many human psychiatric disorders, including schizophrenia. Therefore, in this study, we investigated whether antipsychotic drugs (clozapine (CLZ) and haloperidol (HAL)) affected the formation of heterodimers of D2–5-HT1A receptors as well as 5-HT1A–5-HT2A receptors. Proximity ligation assay (PLA) was used to accurately visualize, for the first time, GPCR heterodimers both at in vitro and ex vivo levels. In line with our previous behavioral studies, we used ketamine to induce cognitive deficits in mice. Our study confirmed the co-localization of D2/5-HT1A and 5-HT1A/5-HT2A receptors in the mouse cortex. Low-dose CLZ (0.3 mg/kg) administered repeatedly, but not CLZ at 1 mg/kg, increased the level of D2–5-HT1A and 5-HT1A–5-HT2A heterodimers in the mouse prefrontal and frontal cortex. On the other hand, HAL decreased the level of GPCR heterodimers. Ketamine affected the formation of 5-HT1A–5-HT2A, but not D2–5-HT1A, heterodimers.
Collapse
Affiliation(s)
- Marta Szlachta
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Maciej Kuśmider
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Paulina Pabian
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Joanna Solich
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Magdalena Kolasa
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Dariusz Żurawek
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | | | - Agata Faron-Górecka
- Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
36
|
Chien T, Weng YT, Chang SY, Lai HL, Chiu FL, Kuo HC, Chuang DM, Chern Y. GSK3β negatively regulates TRAX, a scaffold protein implicated in mental disorders, for NHEJ-mediated DNA repair in neurons. Mol Psychiatry 2018; 23:2375-2390. [PMID: 29298990 PMCID: PMC6294740 DOI: 10.1038/s41380-017-0007-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 10/28/2017] [Accepted: 10/30/2017] [Indexed: 12/27/2022]
Abstract
Translin-associated protein X (TRAX) is a scaffold protein with various functions and has been associated with mental illnesses, including schizophrenia. We have previously demonstrated that TRAX interacts with a Gsα protein-coupled receptor, the A2A adenosine receptor (A2AR), and mediates the function of this receptor in neuritogenesis. In addition, stimulation of the A2AR markedly ameliorates DNA damage evoked by elevated oxidative stress in neurons derived from induced pluripotent stem cells (iPSCs). Here, we report that glycogen synthase kinase 3 beta (GSK3β) and disrupted-in-schizophrenia 1 (DISC1) are two novel interacting proteins of TRAX. We present evidence to suggest that the stimulation of A2AR markedly facilitated DNA repair through the TRAX/DISC1/GSK3β complex in a rat neuronal cell line (PC12), primary mouse neurons, and human medium spiny neurons derived from iPSCs. A2AR stimulation led to the inhibition of GSK3β, thus dissociating the TRAX/DISC1/GSK3β complex and facilitating the non-homologous end-joining pathway (NHEJ) by enhancing the activation of a DNA-dependent protein kinase via phosphorylation at Thr2609. Similarly, pharmacological inhibition of GSK3β by SB216763 also facilitated the TRAX-mediated repair of oxidative DNA damage. Collectively, GSK3β binds with TRAX and negatively affects its ability to facilitate NHEJ repair. The suppression of GSK3β by A2AR activation or a GSK3β inhibitor releases TRAX for the repair of oxidative DNA damage. Our findings shed new light on the molecular mechanisms underlying diseases associated with DNA damage and provides a novel target (i.e., the TRAX/DISC1/GSK3β complex) for future therapeutic development for mental disorders.
Collapse
Affiliation(s)
- Ting Chien
- 0000 0004 0634 0356grid.260565.2Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan ,0000 0004 0633 7958grid.482251.8Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yu-Ting Weng
- 0000 0004 0633 7958grid.482251.8Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan ,0000 0001 2287 1366grid.28665.3fProgram in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Shu-Yung Chang
- 0000 0004 0633 7958grid.482251.8Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan ,0000 0001 0425 5914grid.260770.4Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan
| | - Hsing-Lin Lai
- 0000 0004 0633 7958grid.482251.8Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Feng-Lan Chiu
- 0000 0001 2287 1366grid.28665.3fInstitute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Hung-Chih Kuo
- 0000 0001 2287 1366grid.28665.3fInstitute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - De-Maw Chuang
- 0000 0004 0464 0574grid.416868.5Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD USA
| | - Yijuang Chern
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan. .,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan. .,Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan. .,Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
37
|
Mach RH. Small Molecule Receptor Ligands for PET Studies of the Central Nervous System-Focus on G Protein Coupled Receptors. Semin Nucl Med 2017; 47:524-535. [PMID: 28826524 DOI: 10.1053/j.semnuclmed.2017.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
G protein-coupled receptors (GPRCs) are a class of proteins that are expressed in high abundance and are responsible for numerous signal transduction pathways in the central nervous system. Consequently, alterations in GPRC function have been associated with a wide variety of neurologic and neuropsychiatric disorders. The development of PET probes for imaging GPRCs has served as a major emphasis of PET radiotracer development and PET imaging studies over the past 30 years. In this review, a basic description of the biology of G proteins and GPRCs is provided. This includes recent evidence of the existence of dimeric and multimeric species of GPRCs that have been termed "receptor mosaics," with an emphasis on the different GPRCs that form complexes with the dopamine D2 receptor. An overview of the different PET radiotracers for imaging the component GPRC within these different multimeric complexes of the D2 receptor is also provided.
Collapse
Affiliation(s)
- Robert H Mach
- Department of Radiology, Perelman School Medicine, University of Pennsylvania, Philadelphia, PA.
| |
Collapse
|
38
|
Scarduzio M, Zimmerman CN, Jaunarajs KL, Wang Q, Standaert DG, McMahon LL. Strength of cholinergic tone dictates the polarity of dopamine D2 receptor modulation of striatal cholinergic interneuron excitability in DYT1 dystonia. Exp Neurol 2017; 295:162-175. [PMID: 28587876 DOI: 10.1016/j.expneurol.2017.06.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 06/01/2017] [Accepted: 06/02/2017] [Indexed: 12/11/2022]
Abstract
Balance between cholinergic and dopaminergic signaling is central to striatal control of movement and cognition. In dystonia, a common disorder of movement, anticholinergic therapy is often beneficial. This observation suggests there is a pathological increase in cholinergic tone, yet direct confirmation is lacking. In DYT1, an early-onset genetic form of dystonia caused by a mutation in the protein torsinA (TorA), the suspected heightened cholinergic tone is commonly attributed to faulty dopamine D2 receptor (D2R) signaling where D2R agonists cause excitation of striatal cholinergic interneurons (ChIs), rather than the normal inhibition of firing observed in wild-type animals, an effect known as "paradoxical excitation". Here, we provide for the first time direct measurement of elevated striatal extracellular acetylcholine (ACh) in a knock-in mouse model of human DYT1 dystonia (TorA∆E/+ mice), confirming a striatal hypercholinergic state. We hypothesized that this elevated extracellular ACh might cause chronic over-activation of muscarinic acetylcholine receptors (mAChRs) and disrupt normal D2R function due to their shared coupling to Gi/o-proteins. We tested this concept in vitro first using a broad-spectrum mAChR antagonist, and then using a M2/M4 mAChR selective antagonist to specifically target mAChRs expressed by ChIs. Remarkably, we found that mAChR inhibition reverses the D2R-mediated paradoxical excitation of ChIs recorded in slices from TorA∆E/+ mice to a typical inhibitory response. Furthermore, we recapitulated the paradoxical D2R excitation of ChIs in striatal slices from wild-type mice within minutes by simply increasing cholinergic tone through pharmacological inhibition of acetylcholinesterase (AChE) or by prolonged agonist activation of mAChRs. Collectively, these results show that enhanced mAChR tone itself is sufficient to rapidly reverse the polarity of D2R regulation of ChI excitability, correcting the previous notion that the D2R mediated paradoxical ChI excitation causes the hypercholinergic state in dystonia. Further, using a combination of genetic and pharmacological approaches, we found evidence that this switch in D2R polarity results from a change in coupling from the preferred Gi/o pathway to non-canonical β-arrestin signaling. These results highlight the need to fully understand how the mutation in TorA leads to pathologically heightened extracellular ACh. Furthermore the discovery of this novel ACh-dopamine interaction and the participation of β-arrestin in regulation of cholinergic interneurons is likely important for other basal ganglia disorders characterized by perturbation of ACh-dopamine balance, including Parkinson and Huntington diseases, l-DOPA-induced dyskinesia and schizophrenia.
Collapse
Affiliation(s)
- Mariangela Scarduzio
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Chelsea N Zimmerman
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Karen L Jaunarajs
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Qin Wang
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - David G Standaert
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lori L McMahon
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
39
|
Differences in 5-HT2A and mGlu2 Receptor Expression Levels and Repressive Epigenetic Modifications at the 5-HT2A Promoter Region in the Roman Low- (RLA-I) and High- (RHA-I) Avoidance Rat Strains. Mol Neurobiol 2017; 55:1998-2012. [PMID: 28265857 DOI: 10.1007/s12035-017-0457-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 02/13/2017] [Indexed: 01/12/2023]
Abstract
The serotonin 2A (5-HT2A) and metabotropic glutamate 2 (mGlu2) receptors regulate each other and are associated with schizophrenia. The Roman high- (RHA-I) and the Roman low- (RLA-I) avoidance rat strains present well-differentiated behavioral profiles, with the RHA-I strain emerging as a putative genetic rat model of schizophrenia-related features. The RHA-I strain shows increased 5-HT2A and decreased mGlu2 receptor binding levels in prefrontal cortex (PFC). Here, we looked for differences in gene expression and transcriptional regulation of these receptors. The striatum (STR) was included in the analysis. 5-HT2A, 5-HT1A, and mGlu2 mRNA and [3H]ketanserin binding levels were measured in brain homogenates. As expected, 5-HT2A binding was significantly increased in PFC in the RHA-I rats, while no difference in binding was observed in STR. Surprisingly, 5-HT2A gene expression was unchanged in PFC but significantly decreased in STR. mGlu2 receptor gene expression was significantly decreased in both PFC and STR. No differences were observed for the 5-HT1A receptor. Chromatin immunoprecipitation assay revealed increased trimethylation of histone 3 at lysine 27 (H3K27me3) at the promoter region of the HTR2A gene in the STR. We further looked at the Akt/GSK3 signaling pathway, a downstream point of convergence of the serotonin and glutamate system, and found increased phosphorylation levels of GSK3β at tyrosine 216 and increased β-catenin levels in the PFC of the RHA-I rats. These results reveal region-specific regulation of the 5-HT2A receptor in the RHA-I rats probably due to absence of mGlu2 receptor that may result in differential regulation of downstream pathways.
Collapse
|
40
|
Borroto-Escuela DO, Fuxe K. Diversity and bias through dopamine D2R heteroreceptor complexes. Curr Opin Pharmacol 2017; 32:16-22. [DOI: 10.1016/j.coph.2016.10.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/13/2016] [Accepted: 10/14/2016] [Indexed: 12/13/2022]
|
41
|
Borroto-Escuela DO, Tarakanov AO, Bechter K, Fuxe K. IL1R2, CCR2, and CXCR4 May Form Heteroreceptor Complexes with NMDAR and D2R: Relevance for Schizophrenia. Front Psychiatry 2017; 8:24. [PMID: 28261115 PMCID: PMC5309215 DOI: 10.3389/fpsyt.2017.00024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/31/2017] [Indexed: 12/18/2022] Open
Abstract
The mild neuroinflammation hypothesis of schizophrenia was introduced by Bechter in 2001. It has been hypothesized that a hypofunction of glutamatergic signaling via N-methyl-D-aspartate receptors (NMDARs) and hyperactivation of dopamine D2 receptors play a role in schizophrenia. The triplet puzzle theory states that sets of triplet amino acid homologies guide two different receptors toward each other and contributes to the formation of a receptor heteromer. It is, therefore, proposed that putative NMDAR-C-C chemokine receptor type 2 (CCR2), NMDAR-C-X-C chemokine receptor type 4 (CXCR4), and NMDAR- interleukin 1 receptor type II (IL1R2) heteromers can be formed in the neuronal networks in mild neuroinflammation due to demonstration of Gly-Leu-Leu (GLL), Val-Ser-Thr (VST), and/or Ser-Val-Ser (SVS) amino acid homologies between these receptor protomers. This molecular process may underlie the ability to produce symptoms of schizophrenia in mild neuroinflammation. In this state, volume transmission (VT) is increased involving increased extracellular vesicle-mediated VT from microglia and astroglia. These vesicles may contain CCR2, CXCR4, and/or IL1R2 as well as their ligands and upon internalization by endocytic pathways into neurons can form heteroreceptor complexes with NMDAR in the plasma membrane with pathological allosteric receptor-receptor interactions involving increased internalization and reduced NMDAR signaling. The triplet puzzle theory also suggests the formation of putative D2R-CCR2, D2R-CXCR4, and D2R-IL1R2 heteromers in mild neuroinflammation in view of their demonstrated sets of Leu-Tyr-Ser (LYS), Leu-Pro-Phe (LPF), and/or Ser-Leu-Ala (SLA) triplet homologies. These D2R heteroreceptor complexes may also contribute to schizophrenia-like symptoms in mild neuroinflammation by enhancing D2R protomer function.
Collapse
Affiliation(s)
- Dasiel O Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Biomolecular Science, Section of Physiology, Campus Scientifico Enrico Mattei, University of Urbino, Urbino, Italy; Observatorio Cubano de Neurociencias, Grupo Bohío-Estudio, Yaguajay, Cuba
| | - Alexander O Tarakanov
- Russian Academy of Sciences, St. Petersburg Institute for Informatics and Automation , Saint Petersburg , Russia
| | - Karl Bechter
- Clinic for Psychiatry and Psychotherapy II, Ulm University, Bezirkskrankenhaus Günzburg , Günzburg , Germany
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet , Stockholm , Sweden
| |
Collapse
|