1
|
Alachkar A, Phan A, Dabbous T, Alhassen S, Alhassen W, Reynolds B, Rubinstein M, Ferré S, Civelli O. Humanized dopamine D 4.7 receptor male mice display risk-taking behavior and deficits of social recognition and working memory in light/dark-dependent manner. J Neurosci Res 2024; 102:e25299. [PMID: 38361407 PMCID: PMC11503891 DOI: 10.1002/jnr.25299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 01/03/2024] [Accepted: 01/14/2024] [Indexed: 02/17/2024]
Abstract
The dopamine D4 receptor 7-repeat allele (D4.7 R) has been linked with psychiatric disorders such as attention-deficit-hyperactivity disorder, autism, and schizophrenia. However, the highly diverse study populations and often contradictory findings make it difficult to draw reliable conclusions. The D4.7 R has the potential to explain individual differences in behavior. However, there is still a great deal of ambiguity surrounding whether it is causally connected to the etiology of psychiatric disorders. Therefore, humanized D4.7 R mice, with the long third intracellular domain of the human D4.7 R, may provide a valuable tool to examine the relationship between the D4.7 R variant and specific behavioral phenotypes. We report that D4.7 R male mice carrying the humanized D4.7 R variant exhibit distinct behavioral features that are dependent on the light-dark cycle. The behavioral phenotype was characterized by a working memory deficit, delayed decision execution in the light phase, decreased stress and anxiety, and increased risk behavior in the dark phase. Further, D4.7 R mice displayed impaired social recognition memory in both the light and dark phases. These findings provide insight into the potential causal relationship between the human D4.7 R variant and specific behaviors and encourage further consideration of dopamine D4 receptor (DRD4) ligands as novel treatments for psychiatric disorders in which D4.7 R has been implicated.
Collapse
Affiliation(s)
- Amal Alachkar
- Departments of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California-Irvine, CA 92697, USA
- UC Irvine Center for the Neurobiology of Learning and Memory, University of California-Irvine, Irvine, CA 92697, USA
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California-Irvine, CA 92697, USA
| | - Alvin Phan
- Departments of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California-Irvine, CA 92697, USA
| | - Travis Dabbous
- Departments of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California-Irvine, CA 92697, USA
| | - Sammy Alhassen
- Departments of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California-Irvine, CA 92697, USA
| | - Wedad Alhassen
- Departments of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California-Irvine, CA 92697, USA
| | - Bryan Reynolds
- Department of Drama, School of the Arts, University of California-Irvine, CA 92697, USA
| | - Marcelo Rubinstein
- Departamento de Fisiología y Biología Molecular y Celular, Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Buenos Aires, Argentina
| | - Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Olivier Civelli
- Departments of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California-Irvine, CA 92697, USA
- Department of Developmental and Cell Biology, School of Biological Sciences, University of California-Irvine, CA 92697, USA
| |
Collapse
|
2
|
Homar-Ruano P, Cai NS, Casadó-Anguera V, Casadó V, Ferré S, Moreno E, Canela EI. Significant Functional Differences Between Dopamine D 4 Receptor Polymorphic Variants Upon Heteromerization with α 1A Adrenoreceptors. Mol Neurobiol 2023; 60:6566-6583. [PMID: 37464153 PMCID: PMC10533593 DOI: 10.1007/s12035-023-03476-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/30/2023] [Indexed: 07/20/2023]
Abstract
The functional role of the dopamine D4 receptor (D4R) and its main polymorphic variants has become more evident with the demonstration of heteromers of D4R that control the function of frontal cortico-striatal neurons. Those include heteromers with the α2A adrenoceptor (α2AR) and with the D2R, localized in their cortical somato-dendritic region and striatal nerve terminals, respectively. By using biophysical and cell-signaling methods and heteromer-disrupting peptides in mammalian transfected cells and rat brain slice preparations, here we provide evidence for a new functionally relevant D4R heteromer, the α1AR-D4R heteromer, which is also preferentially localized in cortico-striatal glutamatergic terminals. Significant differences in allosteric modulations between heteromers of α1AR with the D4.4R and D4.7R polymorphic variants could be evidenced with the analysis of G protein-dependent and independent signaling. Similar negative allosteric modulations between α1AR and D4R ligands could be demonstrated for both α1AR-D4.4R and α1AR-D4.7R heteromers on G protein-independent signaling, but only for α1AR-D4.4R on G protein-dependent signaling. From these functional differences, it is proposed that the D4.4R variant provides a gain of function of the α1AR-mediated noradrenergic stimulatory control of cortico-striatal glutamatergic neurotransmission, which could result in a decrease in the vulnerability for impulse control-related neuropsychiatric disorders and increase in the vulnerability for posttraumatic stress disorder.
Collapse
Affiliation(s)
- Patricia Homar-Ruano
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, 08028, Spain
| | - Ning-Sheng Cai
- National Institute On Drug Abuse, Intramural Research Program, National Institutes of Health, Department of Health and Human Services, Baltimore, MD, 21224, USA
| | - Verònica Casadó-Anguera
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, 08028, Spain
| | - Vicent Casadó
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, 08028, Spain
| | - Sergi Ferré
- National Institute On Drug Abuse, Intramural Research Program, National Institutes of Health, Department of Health and Human Services, Baltimore, MD, 21224, USA
| | - Estefanía Moreno
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain.
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, 08028, Spain.
| | - Enric I Canela
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, 08028, Spain
| |
Collapse
|
3
|
Kuś J, Saramowicz K, Czerniawska M, Wiese W, Siwecka N, Rozpędek-Kamińska W, Kucharska-Lusina A, Strzelecki D, Majsterek I. Molecular Mechanisms Underlying NMDARs Dysfunction and Their Role in ADHD Pathogenesis. Int J Mol Sci 2023; 24:12983. [PMID: 37629164 PMCID: PMC10454781 DOI: 10.3390/ijms241612983] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is one of the most common neurodevelopmental disorders, although the aetiology of ADHD is not yet understood. One proposed theory for developing ADHD is N-methyl-D-aspartate receptors (NMDARs) dysfunction. NMDARs are involved in regulating synaptic plasticity and memory function in the brain. Abnormal expression or polymorphism of some genes associated with ADHD results in NMDAR dysfunction. Correspondingly, NMDAR malfunction in animal models results in ADHD-like symptoms, such as impulsivity and hyperactivity. Currently, there are no drugs for ADHD that specifically target NMDARs. However, NMDAR-stabilizing drugs have shown promise in improving ADHD symptoms with fewer side effects than the currently most widely used psychostimulant in ADHD treatment, methylphenidate. In this review, we outline the molecular and genetic basis of NMDAR malfunction and how it affects the course of ADHD. We also present new therapeutic options related to treating ADHD by targeting NMDAR.
Collapse
Affiliation(s)
- Justyna Kuś
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Kamil Saramowicz
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Maria Czerniawska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Wojciech Wiese
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Natalia Siwecka
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Wioletta Rozpędek-Kamińska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Aleksandra Kucharska-Lusina
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Dominik Strzelecki
- Department of Affective and Psychotic Disorders, Medical University of Lodz, Czechoslowacka 8/10, 92-216 Lodz, Poland;
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| |
Collapse
|
4
|
Ferré S, Sarasola LI, Quiroz C, Ciruela F. Presynaptic adenosine receptor heteromers as key modulators of glutamatergic and dopaminergic neurotransmission in the striatum. Neuropharmacology 2023; 223:109329. [PMID: 36375695 DOI: 10.1016/j.neuropharm.2022.109329] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/12/2022]
Abstract
Adenosine plays a very significant role in modulating striatal glutamatergic and dopaminergic neurotransmission. In the present essay we first review the extensive evidence that indicates this modulation is mediated by adenosine A1 and A2A receptors (A1Rs and A2ARs) differentially expressed by the components of the striatal microcircuit that include cortico-striatal glutamatergic and mesencephalic dopaminergic terminals, and the cholinergic interneuron. This microcircuit mediates the ability of striatal glutamate release to locally promote dopamine release through the intermediate activation of cholinergic interneurons. A1Rs and A2ARs are colocalized in the cortico-striatal glutamatergic terminals, where they form A1R-A2AR and A2AR-cannabinoid CB1 receptor (CB1R) heteromers. We then evaluate recent findings on the unique properties of A1R-A2AR and A2AR-CB1R heteromers, which depend on their different quaternary tetrameric structure. These properties involve different allosteric mechanisms in the two receptor heteromers that provide fine-tune modulation of adenosine and endocannabinoid-mediated striatal glutamate release. Finally, we evaluate the evidence supporting the use of different heteromers containing striatal adenosine receptors as targets for drug development for neuropsychiatric disorders, such as Parkinson's disease and restless legs syndrome, based on the ability or inability of the A2AR to demonstrate constitutive activity in the different heteromers, and the ability of some A2AR ligands to act preferentially as neutral antagonists or inverse agonists, or to have preferential affinity for a specific A2AR heteromer.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, USA.
| | - Laura I Sarasola
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907, L'Hospitalet de Llobregat, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, 08907, L'Hospitalet de Llobregat, Spain
| | - César Quiroz
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, USA
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907, L'Hospitalet de Llobregat, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, 08907, L'Hospitalet de Llobregat, Spain.
| |
Collapse
|
5
|
Burström V, Ågren R, Betari N, Valle-León M, Garro-Martínez E, Ciruela F, Sahlholm K. Dopamine-induced arrestin recruitment and desensitization of the dopamine D4 receptor is regulated by G protein-coupled receptor kinase-2. Front Pharmacol 2023; 14:1087171. [PMID: 36778010 PMCID: PMC9911804 DOI: 10.3389/fphar.2023.1087171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/18/2023] [Indexed: 01/28/2023] Open
Abstract
The dopamine D4 receptor (D4R) is expressed in the retina, prefrontal cortex, and autonomic nervous system and has been implicated in attention deficit hyperactivity disorder (ADHD), substance use disorders, and erectile dysfunction. D4R has also been investigated as a target for antipsychotics due to its high affinity for clozapine. As opposed to the closely related dopamine D2 receptor (D2R), dopamine-induced arrestin recruitment and desensitization at the D4R have not been studied in detail. Indeed, some earlier investigations could not detect arrestin recruitment and desensitization of this receptor upon its activation by agonist. Here, we used a novel nanoluciferase complementation assay to study dopamine-induced recruitment of β-arrestin2 (βarr2; also known as arrestin3) and G protein-coupled receptor kinase-2 (GRK2) to the D4R in HEK293T cells. We also studied desensitization of D4R-evoked G protein-coupled inward rectifier potassium (GIRK; also known as Kir3) current responses in Xenopus oocytes. Furthermore, the effect of coexpression of GRK2 on βarr2 recruitment and GIRK response desensitization was examined. The results suggest that coexpression of GRK2 enhanced the potency of dopamine to induce βarr2 recruitment to the D4R and accelerated the rate of desensitization of D4R-evoked GIRK responses. The present study reveals new details about the regulation of arrestin recruitment to the D4R and thus increases our understanding of the signaling and desensitization of this receptor.
Collapse
Affiliation(s)
- Viktor Burström
- Department of Integrative Medical Biology, Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Richard Ågren
- Department of Neuroscience, Karolinska Institutet, Solna, Sweden
| | - Nibal Betari
- Department of Integrative Medical Biology, Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Marta Valle-León
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain,Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, Barcelona, Spain
| | - Emilio Garro-Martínez
- Department of Integrative Medical Biology, Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain,Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, Barcelona, Spain
| | - Kristoffer Sahlholm
- Department of Integrative Medical Biology, Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden,Department of Neuroscience, Karolinska Institutet, Solna, Sweden,Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, Barcelona, Spain,Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, Barcelona, Spain,*Correspondence: Kristoffer Sahlholm,
| |
Collapse
|
6
|
Silvani A, Ghorayeb I, Manconi M, Li Y, Clemens S. Putative Animal Models of Restless Legs Syndrome: A Systematic Review and Evaluation of Their Face and Construct Validity. Neurotherapeutics 2023; 20:154-178. [PMID: 36536233 PMCID: PMC10119375 DOI: 10.1007/s13311-022-01334-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
Restless legs syndrome (RLS) is a sensorimotor disorder that severely affects sleep. It is characterized by an urge to move the legs, which is often accompanied by periodic limb movements during sleep. RLS has a high prevalence in the population and is usually a life-long condition. While its origins remain unclear, RLS is initially highly responsive to treatment with dopaminergic agonists that target D2-like receptors, in particular D2 and D3, but the long-term response is often unsatisfactory. Over the years, several putative animal models for RLS have been developed, mainly based on the epidemiological and neurochemical link with iron deficiency, treatment efficacy of D2-like dopaminergic agonists, or genome-wide association studies that identified risk factors in the patient population. Here, we present the first systematic review of putative animal models of RLS, provide information about their face and construct validity, and report their role in deciphering the underlying pathophysiological mechanisms that may cause or contribute to RLS. We propose that identifying the causal links between genetic risk factors, altered organ functions, and changes to molecular pathways in neural circuitry will eventually lead to more effective new treatment options that bypass the side effects of the currently used therapeutics in RLS, especially for long-term therapy.
Collapse
Affiliation(s)
- Alessandro Silvani
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum - University of Bologna, Ravenna Campus, Ravenna, Italy
| | - Imad Ghorayeb
- Département de Neurophysiologie Clinique, Pôle Neurosciences Cliniques, CHU de Bordeaux, Bordeaux, France
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, UMR 5287, Université de Bordeaux, Bordeaux, France
- Institut de Neurosciences Cognitives et Intégratives d'Aquitaine, UMR 5287, CNRS, Bordeaux, France
| | - Mauro Manconi
- Sleep Medicine Unit, Neurocenter of Southern Switzerland, EOC, Ospedale Civico, Lugano, Switzerland
- Department of Neurology, University Hospital, Inselspital, Bern, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Yuqing Li
- Department of Neurology, College of Medicine, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Stefan Clemens
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
7
|
Sarasola LI, del Torrent CL, Pérez-Arévalo A, Argerich J, Casajuana-Martín N, Chevigné A, Fernández-Dueñas V, Ferré S, Pardo L, Ciruela F. The ADORA1 mutation linked to early-onset Parkinson’s disease alters adenosine A1-A2A receptor heteromer formation and function. Biomed Pharmacother 2022; 156:113896. [DOI: 10.1016/j.biopha.2022.113896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/17/2022] [Accepted: 10/17/2022] [Indexed: 01/14/2023] Open
|
8
|
Earley CJ, Jones BC, Ferré S. Brain-iron deficiency models of restless legs syndrome. Exp Neurol 2022; 356:114158. [PMID: 35779614 PMCID: PMC9357217 DOI: 10.1016/j.expneurol.2022.114158] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 11/04/2022]
Abstract
Restless legs syndrome (RLS) is a common sensorimotor disorder for which two main pathological elements are fairly well accepted: Brain iron deficiency (BID) and an altered dopaminergic system. The ability to better understand the causal and consequential factors related to these two pathological elements, would hopefully lead to the development of better therapeutic strategies for treating, if not curing, this disease. The current understanding of the relationship between these two elements is that BID leads to some alterations in neurotransmitters and subsequent changes in the dopaminergic system. Therefore, rodent models based on diet-induced BID, provide a biological substrate to understand the consequences of BID on dopaminergic pathway and on alternative pathways that may be involved. In this review, we present the current research on dopaminergic changes found in RLS subjects and compare that to what is seen in the BID rodent model to provide a validation of the BID rodent model. We also demonstrate the ability of the BID model to predict changes in other neurotransmitter systems and how that has led to new treatment options. Finally, we will present arguments for the utility of recombinant inbred mouse strains that demonstrate natural variation in brain iron, to explore the genetic basis of altered brain iron homeostasis as a model to understand why in idiopathic RLS there can exist a BID despite normal peripheral iron store. This review is the first to draw on 25 years of human and basic research into the pathophysiology of RLS to provide strong supportive data as to the validity of BID model as an important translational model of the disease. As we will demonstrate here, not only does the BID model closely and accurately mimic what we see in the dopaminergic system of RLS, it is the first model to identify alternative systems from which new treatments have recently been developed.
Collapse
Affiliation(s)
- Christopher J Earley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Byron C Jones
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sergi Ferré
- Integrative Neurobiology Section, National Institutes of Health/National Institute on Drug Abuse, Baltimore, MD, USA
| |
Collapse
|
9
|
Kessi M, Duan H, Xiong J, Chen B, He F, Yang L, Ma Y, Bamgbade OA, Peng J, Yin F. Attention-deficit/hyperactive disorder updates. Front Mol Neurosci 2022; 15:925049. [PMID: 36211978 PMCID: PMC9532551 DOI: 10.3389/fnmol.2022.925049] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 08/26/2022] [Indexed: 11/15/2022] Open
Abstract
Background Attention-deficit/hyperactive disorder (ADHD) is a neurodevelopmental disorder that commonly occurs in children with a prevalence ranging from 3.4 to 7.2%. It profoundly affects academic achievement, well-being, and social interactions. As a result, this disorder is of high cost to both individuals and society. Despite the availability of knowledge regarding the mechanisms of ADHD, the pathogenesis is not clear, hence, the existence of many challenges especially in making correct early diagnosis and provision of accurate management. Objectives We aimed to review the pathogenic pathways of ADHD in children. The major focus was to provide an update on the reported etiologies in humans, animal models, modulators, therapies, mechanisms, epigenetic changes, and the interaction between genetic and environmental factors. Methods References for this review were identified through a systematic search in PubMed by using special keywords for all years until January 2022. Results Several genes have been reported to associate with ADHD: DRD1, DRD2, DRD4, DAT1, TPH2, HTR1A, HTR1B, SLC6A4, HTR2A, DBH, NET1, ADRA2A, ADRA2C, CHRNA4, CHRNA7, GAD1, GRM1, GRM5, GRM7, GRM8, TARBP1, ADGRL3, FGF1, MAOA, BDNF, SNAP25, STX1A, ATXN7, and SORCS2. Some of these genes have evidence both from human beings and animal models, while others have evidence in either humans or animal models only. Notably, most of these animal models are knockout and do not generate the genetic alteration of the patients. Besides, some of the gene polymorphisms reported differ according to the ethnic groups. The majority of the available animal models are related to the dopaminergic pathway. Epigenetic changes including SUMOylation, methylation, and acetylation have been reported in genes related to the dopaminergic pathway. Conclusion The dopaminergic pathway remains to be crucial in the pathogenesis of ADHD. It can be affected by environmental factors and other pathways. Nevertheless, it is still unclear how environmental factors relate to all neurotransmitter pathways; thus, more studies are needed. Although several genes have been related to ADHD, there are few animal model studies on the majority of the genes, and they do not generate the genetic alteration of the patients. More animal models and epigenetic studies are required.
Collapse
Affiliation(s)
- Miriam Kessi
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Haolin Duan
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Juan Xiong
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Baiyu Chen
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Fang He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Lifen Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Yanli Ma
- Department of Neurology, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Olumuyiwa A. Bamgbade
- Department of Anesthesiology and Pharmacology, University of British Columbia, Vancouver, BC, Canada
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
- *Correspondence: Fei Yin,
| |
Collapse
|
10
|
Lindenbach D, Vacca G, Ahn S, Seamans JK, Phillips AG. Optogenetic modulation of glutamatergic afferents from the ventral subiculum to the nucleus accumbens: Effects on dopamine function, response vigor and locomotor activity. Behav Brain Res 2022; 434:114028. [DOI: 10.1016/j.bbr.2022.114028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/13/2022] [Accepted: 07/25/2022] [Indexed: 01/06/2023]
|
11
|
Kanarik M, Grimm O, Mota NR, Reif A, Harro J. ADHD co-morbidities: A review of implication of gene × environment effects with dopamine-related genes. Neurosci Biobehav Rev 2022; 139:104757. [PMID: 35777579 DOI: 10.1016/j.neubiorev.2022.104757] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 02/07/2023]
Abstract
ADHD is a major burden in adulthood, where co-morbid conditions such as depression, substance use disorder and obesity often dominate the clinical picture. ADHD has substantial shared heritability with other mental disorders, contributing to comorbidity. However, environmental risk factors exist but their interaction with genetic makeup, especially in relation to comorbid disorders, remains elusive. This review for the first time summarizes present knowledge on gene x environment (GxE) interactions regarding the dopamine system. Hitherto, mainly candidate (GxE) studies were performed, focusing on the genes DRD4, DAT1 and MAOA. Some evidence suggest that the variable number tandem repeats in DRD4 and MAOA may mediate GxE interactions in ADHD generally, and comorbid conditions specifically. Nevertheless, even for these genes, common variants are bound to suggest risk only in the context of gender and specific environments. For other polymorphisms, evidence is contradictory and less convincing. Particularly lacking are longitudinal studies testing the interaction of well-defined environmental with polygenic risk scores reflecting the dopamine system in its entirety.
Collapse
Affiliation(s)
- Margus Kanarik
- Chair of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Ravila 14A Chemicum, 50411 Tartu, Estonia
| | - Oliver Grimm
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Nina Roth Mota
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Psychiatry, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Jaanus Harro
- Chair of Neuropsychopharmacology, Institute of Chemistry, University of Tartu, Ravila 14A Chemicum, 50411 Tartu, Estonia; Psychiatry Clinic, North Estonia Medical Centre, Paldiski Road 52, 10614 Tallinn, Estonia.
| |
Collapse
|
12
|
Casadó-Anguera V, Casadó V. Unmasking allosteric binding sites: Novel targets for GPCR drug discovery. Expert Opin Drug Discov 2022; 17:897-923. [PMID: 35649692 DOI: 10.1080/17460441.2022.2085684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Unexpected non-apparent and hidden allosteric binding sites are non-classical and non-apparent allosteric centers in 3-D X-ray protein structures until orthosteric or allosteric ligands bind to them. The orthosteric center of one protomer that modulates binding centers of the other protomers within an oligomer is also an unexpected allosteric site. Furthermore, another partner protein can also produce these effects, acting as an unexpected allosteric modulator. AREAS COVERED This review summarizes both classical and non-classical allosterism. The authors focus on G protein-coupled receptor (GPCR) oligomers as a paradigm of allosteric molecules. Moreover, they show several examples of unexpected allosteric sites such as hidden allosteric sites in a protomer that appear after the interaction with other molecules and the allosterism exerted between orthosteric sites within GPCR oligomer, emphasizing on the allosteric modulations that can occur between binding sites. EXPERT OPINION The study of these new non-classical allosteric sites will expand the diversity of allosteric control on the function of orthosteric sites within proteins, whether GPCRs or other receptors, enzymes or transporters. Moreover, the design of new drugs targeting these hidden allosteric sites or already known orthosteric sites acting as allosteric sites in protein homo- or hetero-oligomers will increase the therapeutic potential of allosterism.
Collapse
Affiliation(s)
- Verònica Casadó-Anguera
- Laboratory of Molecular Neuropharmacology, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, and Institute of Biomedicine of the Universitat de Barcelona, Barcelona, Spain.,Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Vicent Casadó
- Laboratory of Molecular Neuropharmacology, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, and Institute of Biomedicine of the Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
13
|
Rodrigues MS, Ferreira SG, Quiroz C, Earley CJ, García-Borreguero D, Cunha RA, Ciruela F, Köfalvi A, Ferré S. Brain Iron Deficiency Changes the Stoichiometry of Adenosine Receptor Subtypes in Cortico-Striatal Terminals: Implications for Restless Legs Syndrome. Molecules 2022; 27:1489. [PMID: 35268590 PMCID: PMC8911604 DOI: 10.3390/molecules27051489] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 01/01/2023] Open
Abstract
Brain iron deficiency (BID) constitutes a primary pathophysiological mechanism in restless legs syndrome (RLS). BID in rodents has been widely used as an animal model of RLS, since it recapitulates key neurochemical changes reported in RLS patients and shows an RLS-like behavioral phenotype. Previous studies with the BID-rodent model of RLS demonstrated increased sensitivity of cortical pyramidal cells to release glutamate from their striatal nerve terminals driving striatal circuits, a correlative finding of the cortical motor hyperexcitability of RLS patients. It was also found that BID in rodents leads to changes in the adenosinergic system, a downregulation of the inhibitory adenosine A1 receptors (A1Rs) and upregulation of the excitatory adenosine A2A receptors (A2ARs). It was then hypothesized, but not proven, that the BID-induced increased sensitivity of cortico-striatal glutamatergic terminals could be induced by a change in A1R/A2AR stoichiometry in favor of A2ARs. Here, we used a newly developed FACS-based synaptometric analysis to compare the relative abundance on A1Rs and A2ARs in cortico-striatal and thalamo-striatal glutamatergic terminals (labeled with vesicular glutamate transporters VGLUT1 and VGLUT2, respectively) of control and BID rats. It could be demonstrated that BID (determined by measuring transferrin receptor density in the brain) is associated with a selective decrease in the A1R/A2AR ratio in VGLUT1 positive-striatal terminals.
Collapse
Affiliation(s)
- Matilde S. Rodrigues
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; (M.S.R.); (S.G.F.); (R.A.C.); (A.K.)
| | - Samira G. Ferreira
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; (M.S.R.); (S.G.F.); (R.A.C.); (A.K.)
| | - César Quiroz
- Integrative Neurobiology Section, National Institute on Drug Abuse, Baltimore, MD 21224, USA;
| | | | | | - Rodrigo A. Cunha
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; (M.S.R.); (S.G.F.); (R.A.C.); (A.K.)
- Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L’Hospitalet de Llobregat, Spain;
- Neuropharmacology and Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Belvitge, Idibell, 08907 L’Hospitalet de Llobregat, Spain
| | - Attila Köfalvi
- CNC-Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, 3004-504 Coimbra, Portugal; (M.S.R.); (S.G.F.); (R.A.C.); (A.K.)
| | - Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Baltimore, MD 21224, USA;
| |
Collapse
|
14
|
Ferré S, Belcher AM, Bonaventura J, Quiroz C, Sánchez-Soto M, Casadó-Anguera V, Cai NS, Moreno E, Boateng CA, Keck TM, Florán B, Earley CJ, Ciruela F, Casadó V, Rubinstein M, Volkow ND. Functional and pharmacological role of the dopamine D 4 receptor and its polymorphic variants. Front Endocrinol (Lausanne) 2022; 13:1014678. [PMID: 36267569 PMCID: PMC9578002 DOI: 10.3389/fendo.2022.1014678] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
The functional and pharmacological significance of the dopamine D4 receptor (D4R) has remained the least well understood of all the dopamine receptor subtypes. Even more enigmatic has been the role of the very prevalent human DRD4 gene polymorphisms in the region that encodes the third intracellular loop of the receptor. The most common polymorphisms encode a D4R with 4 or 7 repeats of a proline-rich sequence of 16 amino acids (D4.4R and D4.7R). DRD4 polymorphisms have been associated with individual differences linked to impulse control-related neuropsychiatric disorders, with the most consistent associations established between the gene encoding D4.7R and attention-deficit hyperactivity disorder (ADHD) and substance use disorders. The function of D4R and its polymorphic variants is being revealed by addressing the role of receptor heteromerization and the relatively avidity of norepinephrine for D4R. We review the evidence conveying a significant and differential role of D4.4R and D4.7R in the dopaminergic and noradrenergic modulation of the frontal cortico-striatal pyramidal neuron, with implications for the moderation of constructs of impulsivity as personality traits. This differential role depends on their ability to confer different properties to adrenergic α2A receptor (α2AR)-D4R heteromers and dopamine D2 receptor (D2R)-D4R heteromers, preferentially localized in the perisomatic region of the frontal cortical pyramidal neuron and its striatal terminals, respectively. We also review the evidence to support the D4R as a therapeutic target for ADHD and other impulse-control disorders, as well as for restless legs syndrome.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, United States
- *Correspondence: Sergi Ferré,
| | - Annabelle M. Belcher
- Division of Addiction Research and Treatment, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jordi Bonaventura
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, United States
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain
| | - César Quiroz
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, United States
| | - Marta Sánchez-Soto
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, United States
| | - Verònica Casadó-Anguera
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| | - Ning-Sheng Cai
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes on Drug Abuse, Baltimore, MD, United States
| | - Estefanía Moreno
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| | - Comfort A. Boateng
- Department of Basic Pharmaceutical Sciences, Fred Wilson School of Pharmacy, High Point, NC, United States
| | - Thomas M. Keck
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ, United States
| | - Benjamín Florán
- Departament of Physiology, Biophysics and Neurosciences, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Christopher J. Earley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, Bellvitge Institute for Biomedical Research, L'Hospitalet de Llobregat, Spain
| | - Vicent Casadó
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona, Spain
| | - Marcelo Rubinstein
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas and, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Nora D. Volkow
- National Institute on Drug Abuse, National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
15
|
Abstract
Restless Legs Syndrome (RLS) is a sensorimotor disorder that severely affects sleep. It is characterized by an urge to move the legs that is often accompanied by periodic limb movements during sleep (PLMS). RLS has a high prevalence in the population and is usually a life-long condition. While its origins remain unclear, RLS is initially highly responsive to treatment with dopaminergics that target the D3 receptor. However, over time patients often develop a gradual tolerance that can lead to the emergence of adverse effects and the augmentation of the symptoms. While the basal ganglia and the striatum control leg movements, the lumbar spinal cord is the gateway for the sensory processing of the symptoms and critical for the associated leg movements. D3 receptors are highly expressed in nucleus accumbens (NAc) of the striatum and the sensory-processing areas of the spinal dorsal horn. In contrast, D1 receptors are strongly expressed throughout the entire striatum and in the ventral horn of the spinal cord. Long-term treatment with D3 receptor full agonists is associated with an upregulation of the D1 receptor subtype, and D3 and D1 receptors can form functional heteromers, in which the D3R controls the D1R function. It is conceivable that the switch from beneficial treatment to augmentation observed in RLS patients after prolonged D3R agonist exposure may be the result of unmasked D1-like receptor actions.
Collapse
Affiliation(s)
- Stefan Clemens
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
16
|
The Role of Iron and Zinc in the Treatment of ADHD among Children and Adolescents: A Systematic Review of Randomized Clinical Trials. Nutrients 2021; 13:nu13114059. [PMID: 34836314 PMCID: PMC8618748 DOI: 10.3390/nu13114059] [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: 09/17/2021] [Revised: 11/01/2021] [Accepted: 11/11/2021] [Indexed: 12/18/2022] Open
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a neurodevelopmental disorder common from childhood to adulthood, affecting 5% to 12% among the general population in developed countries. Potential etiological factors have been identified, including genetic causes, environmental elements and epigenetic components. Nutrition is currently considered an influencing factor, and several studies have explored the contribution of restriction and dietary supplements in ADHD treatments. Iron is an essential cofactor required for a number of functions, such as transport of oxygen, immune function, cellular respiration, neurotransmitter metabolism (dopamine production), and DNA synthesis. Zinc is also an essential trace element, required for cellular functions related to the metabolism of neurotransmitters, melatonin, and prostaglandins. Epidemiological studies have found that iron and zinc deficiencies are common nutritional deficits worldwide, with important roles on neurologic functions (poor memory, inattentiveness, and impulsiveness), finicky appetite, and mood changes (sadness and irritability). Altered levels of iron and zinc have been related with the aggravation and progression of ADHD. Objective: This is a systematic review focused on the contribution of iron and zinc in the progression of ADHD among children and adolescents, and how therapies including these elements are tolerated along with its effectiveness (according to PRISMA guidelines). Method: The scientific literature was screened for randomized controlled trials published between January 2000 to July 2021. The databases consulted were Medline, PsycINFO, Web of Science, and Google Scholar. Two independent reviewers screened studies, extracted data, and assessed quality and risk of bias (CONSORT, NICE, and Cochrane checklists used). Conclusion: Nine studies met the eligibility criteria and were selected. Evidence was obtained regarding the contribution of iron-zinc supplementation in the treatment of ADHD among young individuals. The discussion was focused on how the deficits of these elements contribute to affectation on multiple ADHD correlates, and potential mechanisms explaining the mediational pathways. Evidence also suggested that treating ADHD with diet interventions might be particularly useful for specific subgroups of children and adolescents, but further investigations of the effects of these diet interventions are needed.
Collapse
|
17
|
Synaptic Zn 2+ potentiates the effects of cocaine on striatal dopamine neurotransmission and behavior. Transl Psychiatry 2021; 11:570. [PMID: 34750356 PMCID: PMC8575899 DOI: 10.1038/s41398-021-01693-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
Abstract
Cocaine binds to the dopamine (DA) transporter (DAT) to regulate cocaine reward and seeking behavior. Zinc (Zn2+) also binds to the DAT, but the in vivo relevance of this interaction is unknown. We found that Zn2+ concentrations in postmortem brain (caudate) tissue from humans who died of cocaine overdose were significantly lower than in control subjects. Moreover, the level of striatal Zn2+ content in these subjects negatively correlated with plasma levels of benzoylecgonine, a cocaine metabolite indicative of recent use. In mice, repeated cocaine exposure increased synaptic Zn2+ concentrations in the caudate putamen (CPu) and nucleus accumbens (NAc). Cocaine-induced increases in Zn2+ were dependent on the Zn2+ transporter 3 (ZnT3), a neuronal Zn2+ transporter localized to synaptic vesicle membranes, as ZnT3 knockout (KO) mice were insensitive to cocaine-induced increases in striatal Zn2+. ZnT3 KO mice showed significantly lower electrically evoked DA release and greater DA clearance when exposed to cocaine compared to controls. ZnT3 KO mice also displayed significant reductions in cocaine locomotor sensitization, conditioned place preference (CPP), self-administration, and reinstatement compared to control mice and were insensitive to cocaine-induced increases in striatal DAT binding. Finally, dietary Zn2+ deficiency in mice resulted in decreased striatal Zn2+ content, cocaine locomotor sensitization, CPP, and striatal DAT binding. These results indicate that cocaine increases synaptic Zn2+ release and turnover/metabolism in the striatum, and that synaptically released Zn2+ potentiates the effects of cocaine on striatal DA neurotransmission and behavior and is required for cocaine-primed reinstatement. In sum, these findings reveal new insights into cocaine's pharmacological mechanism of action and suggest that Zn2+ may serve as an environmentally derived regulator of DA neurotransmission, cocaine pharmacodynamics, and vulnerability to cocaine use disorders.
Collapse
|
18
|
Heteromerization between α 2A adrenoceptors and different polymorphic variants of the dopamine D 4 receptor determines pharmacological and functional differences. Implications for impulsive-control disorders. Pharmacol Res 2021; 170:105745. [PMID: 34182128 PMCID: PMC9885860 DOI: 10.1016/j.phrs.2021.105745] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/11/2021] [Accepted: 06/23/2021] [Indexed: 02/01/2023]
Abstract
Polymorphic alleles of the human dopamine D4 receptor gene (DRD4) have been consistently associated with individual differences in personality traits and neuropsychiatric disorders, particularly between the gene encoding dopamine D4.7 receptor variant and attention deficit hyperactivity disorder (ADHD). The α2A adrenoceptor gene has also been associated with ADHD. In fact, drugs targeting the α2A adrenoceptor (α2AR), such as guanfacine, are commonly used in ADHD treatment. In view of the involvement of dopamine D4 receptor (D4R) and α2AR in ADHD and impulsivity, their concurrent localization in cortical pyramidal neurons and the demonstrated ability of D4R to form functional heteromers with other G protein-coupled receptors, in this study we evaluate whether the α2AR forms functional heteromers with D4R and weather these heteromers show different properties depending on the D4R variant involved. Using cortical brain slices from hD4.7R knock-in and wild-type mice, here, we demonstrate that α2AR and D4R heteromerize and constitute a significant functional population of cortical α2AR and D4R. Moreover, in cortical slices from wild-type mice and in cells transfected with α2AR and D4.4R, we detect a negative crosstalk within the heteromer. This negative crosstalk is lost in cortex from hD4.7R knock-in mice and in cells expressing the D4.7R polymorphic variant. We also show a lack of efficacy of D4R ligands to promote G protein activation and signaling only within the α2AR-D4.7R heteromer. Taken together, our results suggest that α2AR-D4R heteromers play a pivotal role in catecholaminergic signaling in the brain cortex and are likely targets for ADHD pharmacotherapy.
Collapse
|
19
|
Ferré S, Guitart X, Quiroz C, Rea W, García-Malo C, Garcia-Borreguero D, Allen RP, Earley CJ. Akathisia and Restless Legs Syndrome: Solving the Dopaminergic Paradox. Sleep Med Clin 2021; 16:249-267. [PMID: 33985651 DOI: 10.1016/j.jsmc.2021.02.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Akathisia is an urgent need to move that is associated with treatment with dopamine receptor blocking agents (DRBAs) and with restless legs syndrome (RLS). The pathogenetic mechanism of akathisia has not been resolved. This article proposes that it involves an increased presynaptic dopaminergic transmission in the ventral striatum and concomitant strong activation of postsynaptic dopamine D1 receptors, which form complexes (heteromers) with dopamine D3 and adenosine A1 receptors. It also proposes that in DRBA-induced akathisia, increased dopamine release depends on inactivation of autoreceptors, whereas in RLS it depends on a brain iron deficiency-induced down-regulation of striatal presynaptic A1 receptors.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Triad Building, 333 Cassell Drive, Baltimore, MD 21224, USA.
| | - Xavier Guitart
- Integrative Neurobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Triad Building, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - César Quiroz
- Integrative Neurobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Triad Building, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - William Rea
- Integrative Neurobiology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Triad Building, 333 Cassell Drive, Baltimore, MD 21224, USA
| | - Celia García-Malo
- Sleep Research Institute, Paseo de la Habana 151, Madrid 28036, Spain
| | | | - Richard P Allen
- Department of Neurology, Johns Hopkins University, Johns Hopkins Bayview Medical Center, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA
| | - Christopher J Earley
- Department of Neurology, Johns Hopkins University, Johns Hopkins Bayview Medical Center, 5501 Hopkins Bayview Circle, Baltimore, MD 21224, USA
| |
Collapse
|
20
|
Rahi V, Kumar P. Animal models of attention-deficit hyperactivity disorder (ADHD). Int J Dev Neurosci 2021; 81:107-124. [PMID: 33428802 DOI: 10.1002/jdn.10089] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/14/2020] [Accepted: 01/06/2021] [Indexed: 12/15/2022] Open
Abstract
Attention-deficit hyperactivity disorder (ADHD) is a heterogeneous neuropsychiatric disorder characterized by three primary symptoms hyperactivity, attention deficit, and impulsiveness, observed in both children and adults. In childhood, this disorder is more common in boys than in girls, and at least 75% will continue to suffer from the disorder until adulthood. Individuals with ADHD generally have poor academic, occupational, and social functioning resulting from developmentally inappropriate levels of hyperactivity and impulsivity, as well as impaired ability to maintain attention on motivationally relevant tasks. Very few drugs available in clinical practice altogether abolish the symptoms of ADHD, therefore, to find new drugs and target it is essential to understand the neuropathological, neurochemical, and genetic alterations that lead to the progression of ADHD. With this contrast, an animal study is the best approach because animal models provide relatively fast invasive manipulation, rigorous hypothesis testing, as well as it provides a better angle to understand the pathological mechanisms involved in disease progression. Moreover, animal models, especially for ADHD, serve with good predictive validity would allow the assessment and development of new therapeutic interventions, with this aim, the present review collect the various animal models on a single platform so that the research can select an appropriate model to pursue his study.
Collapse
Affiliation(s)
- Vikrant Rahi
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, India
| | - Puneet Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, India
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| |
Collapse
|
21
|
Park BY, Bethlehem RAI, Paquola C, Larivière S, Rodríguez-Cruces R, Vos de Wael R, Bullmore ET, Bernhardt BC. An expanding manifold in transmodal regions characterizes adolescent reconfiguration of structural connectome organization. eLife 2021; 10:e64694. [PMID: 33787489 PMCID: PMC8087442 DOI: 10.7554/elife.64694] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 03/30/2021] [Indexed: 12/13/2022] Open
Abstract
Adolescence is a critical time for the continued maturation of brain networks. Here, we assessed structural connectome development in a large longitudinal sample ranging from childhood to young adulthood. By projecting high-dimensional connectomes into compact manifold spaces, we identified a marked expansion of structural connectomes, with strongest effects in transmodal regions during adolescence. Findings reflected increased within-module connectivity together with increased segregation, indicating increasing differentiation of higher-order association networks from the rest of the brain. Projection of subcortico-cortical connectivity patterns into these manifolds showed parallel alterations in pathways centered on the caudate and thalamus. Connectome findings were contextualized via spatial transcriptome association analysis, highlighting genes enriched in cortex, thalamus, and striatum. Statistical learning of cortical and subcortical manifold features at baseline and their maturational change predicted measures of intelligence at follow-up. Our findings demonstrate that connectome manifold learning can bridge the conceptual and empirical gaps between macroscale network reconfigurations, microscale processes, and cognitive outcomes in adolescent development.
Collapse
Affiliation(s)
- Bo-yong Park
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill UniversityMontrealCanada
- Department of Data Science, Inha UniversityIncheonRepublic of Korea
| | - Richard AI Bethlehem
- Autism Research Centre, Department of Psychiatry, University of CambridgeCambridgeUnited Kingdom
- Brain Mapping Unit, Department of Psychiatry, University of CambridgeCambridgeUnited Kingdom
| | - Casey Paquola
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill UniversityMontrealCanada
- Institute of Neuroscience and Medicine (INM-1), Forschungszentrum JülichJülichGermany
| | - Sara Larivière
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill UniversityMontrealCanada
| | - Raul Rodríguez-Cruces
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill UniversityMontrealCanada
| | - Reinder Vos de Wael
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill UniversityMontrealCanada
| | - Edward T Bullmore
- Brain Mapping Unit, Department of Psychiatry, University of CambridgeCambridgeUnited Kingdom
| | - Boris C Bernhardt
- McConnell Brain Imaging Centre, Montreal Neurological Institute and Hospital, McGill UniversityMontrealCanada
| |
Collapse
|
22
|
Giorgioni G, Del Bello F, Pavletić P, Quaglia W, Botticelli L, Cifani C, Micioni Di Bonaventura E, Micioni Di Bonaventura MV, Piergentili A. Recent findings leading to the discovery of selective dopamine D 4 receptor ligands for the treatment of widespread diseases. Eur J Med Chem 2020; 212:113141. [PMID: 33422983 DOI: 10.1016/j.ejmech.2020.113141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/23/2020] [Accepted: 12/24/2020] [Indexed: 12/14/2022]
Abstract
Since its discovery, the dopamine D4 receptor (D4R) has been suggested to be an attractive target for the treatment of neuropsychiatric diseases. Novel findings have renewed the interest in such a receptor as an emerging target for the management of different diseases, including cancer, Parkinson's disease, alcohol or substance use disorders, eating disorders, erectile dysfunction and cognitive deficits. The recently resolved crystal structures of D4R in complexes with the potent ligands nemonapride and L-745870 strongly improved the knowledge on the molecular mechanisms involving the D4R functions and may help medicinal chemists in drug design. This review is focused on the recent development of the subtype selective D4R ligands belonging to classical or new chemotypes. Moreover, ligands showing functional selectivity toward G protein activation or β-arrestin recruitment and the effects of selective D4R ligands on the above-mentioned diseases are discussed.
Collapse
Affiliation(s)
- Gianfabio Giorgioni
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032, Camerino, Italy
| | - Fabio Del Bello
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032, Camerino, Italy.
| | - Pegi Pavletić
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032, Camerino, Italy
| | - Wilma Quaglia
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032, Camerino, Italy.
| | - Luca Botticelli
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna Delle Carceri 9, 62032, Camerino, Italy
| | - Carlo Cifani
- School of Pharmacy, Pharmacology Unit, University of Camerino, Via Madonna Delle Carceri 9, 62032, Camerino, Italy
| | | | | | - Alessandro Piergentili
- School of Pharmacy, Medicinal Chemistry Unit, University of Camerino, Via S. Agostino 1, 62032, Camerino, Italy
| |
Collapse
|
23
|
García-Pedraza JÁ, Morán A, Martín ML, Ollauri-Ibáñez C, Rodríguez-Barbero A, Villalón CM, García-Domingo M. Dopamine D 4 receptor subtype activation reduces the rat cardiac parasympathetic discharge. Pflugers Arch 2020; 472:1693-1703. [PMID: 32820344 DOI: 10.1007/s00424-020-02452-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/31/2020] [Accepted: 08/14/2020] [Indexed: 12/19/2022]
Abstract
The dopaminergic system influences the heart rhythm by inhibiting the rat cardiac sympathetic and parasympathetic neurotransmissions through activation of D2-like receptors (encompassing the D2, D3, and D4 subtypes). Whereas D2 receptor subtype activation results in cardiac sympatho-inhibition, the dopamine receptor subtypes involved in rat cardiac vago-inhibition remain unknown. Hence, this study investigated the specific functional role of the D2-like receptor subtypes (D2, D3, and/or D4) inhibiting the rat heart cholinergic drive. For this purpose, male Wistar rats were pithed and prepared for cardiac vagal stimulation. Bradycardic responses were obtained by electrical stimulation of vagal fibres (3, 6, 9 Hz; n = 100) or i.v. acetylcholine (ACh; 1, 5, 10 μg/kg; n = 15). Expression of D2, D3, and D4 receptors was studied in left and right atrium samples by PCR (n = 4). Intravenous injections of quinpirole (D2-like agonist; 1-30 μg/kg), but not of SFK-38393 (D1-like agonist; 1-30 μg/kg), dose-dependently inhibited the vagally induced bradycardia. The vago-inhibition induced by quinpirole (which failed to affect the bradycardia to i.v. ACh) was unchanged after i.v. injections of the antagonists L-741,626 (D2; 100 μg/kg) or SB-277011-A (D3; 100 μg/kg), but it was abolished by L-745,870 (D4; 100 μg/kg). mRNA levels of D2, D3, and D4 receptor subtype were detected in the left and right rat atria. Our results suggest that the quinpirole-induced vagolytic effect involves prejunctional D4 receptor subtypes, located in the left and right atria. This provides new evidence on the relevance of D4 receptor modulating the heart parasympathetic control.
Collapse
Affiliation(s)
- José Ángel García-Pedraza
- Laboratorio de Farmacología, Dep. de Fisiología y Farmacología, Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, 37007, Salamanca, Spain
| | - Asunción Morán
- Laboratorio de Farmacología, Dep. de Fisiología y Farmacología, Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, 37007, Salamanca, Spain
| | - María Luisa Martín
- Laboratorio de Farmacología, Dep. de Fisiología y Farmacología, Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, 37007, Salamanca, Spain
| | - Claudia Ollauri-Ibáñez
- Unidad de Fisiopatología Renal y Cardiovascular, Instituto Reina Sofía de Investigación Nefrológica, Dep. de Fisiología y Farmacología, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, 37007, Salamanca, Spain
| | - Alicia Rodríguez-Barbero
- Unidad de Fisiopatología Renal y Cardiovascular, Instituto Reina Sofía de Investigación Nefrológica, Dep. de Fisiología y Farmacología, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, 37007, Salamanca, Spain
| | - Carlos M Villalón
- Departamento de Farmacobiología, Cinvestav-Coapa, Czda. Tenorios 235, Col. Granjas-Coapa, Deleg. Tlalpan, 14330, Mexico City, Mexico
| | - Mónica García-Domingo
- Laboratorio de Farmacología, Dep. de Fisiología y Farmacología, Facultad de Farmacia, Instituto de Investigación Biomédica de Salamanca (IBSAL), Universidad de Salamanca, 37007, Salamanca, Spain.
| |
Collapse
|
24
|
Underlying Susceptibility to Eating Disorders and Drug Abuse: Genetic and Pharmacological Aspects of Dopamine D4 Receptors. Nutrients 2020; 12:nu12082288. [PMID: 32751662 PMCID: PMC7468707 DOI: 10.3390/nu12082288] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 07/26/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023] Open
Abstract
The dopamine D4 receptor (DRD4) has a predominant expression in the prefrontal cortex (PFC), brain area strictly involved in the modulation of reward processes related to both food and drug consumption. Additionally, the human DRD4 gene is characterized by a variable number of tandem repeats (VNTR) in the exon 3 and, among the polymorphic variants, the 7-repeat (7R) allele appears as a contributing factor in the neurobiological mechanisms underlying drug abuse, aberrant eating behaviors and related comorbidities. The 7R variant encodes for a receptor with a blunted intracellular response to dopamine, and carriers of this polymorphism might be more tempted to enhance dopamine levels in the brain, through the overconsumption of drugs of abuse or palatable food, considering their reinforcing properties. Moreover, the presence of this polymorphism seems to increase the susceptibility of individuals to engage maladaptive eating patterns in response to negative environmental stimuli. This review is focused on the role of DRD4 and DRD4 genetic polymorphism in these neuropsychiatric disorders in both clinical and preclinical studies. However, further research is needed to better clarify the complex DRD4 role, by using validated preclinical models and novel compounds more selective for DRD4.
Collapse
|
25
|
Knop J, van IJzendoorn MH, Bakermans-Kranenburg MJ, Joëls M, van der Veen R. Maternal care of heterozygous dopamine receptor D4 knockout mice: Differential susceptibility to early-life rearing conditions. GENES BRAIN AND BEHAVIOR 2020; 19:e12655. [PMID: 32306548 PMCID: PMC7540036 DOI: 10.1111/gbb.12655] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/30/2020] [Accepted: 04/13/2020] [Indexed: 12/27/2022]
Abstract
The differential susceptibility hypothesis proposes that individuals who are more susceptible to the negative effects of adverse rearing conditions may also benefit more from enriched environments. Evidence derived from human experiments suggests the lower efficacy dopamine receptor D4 (DRD4) 7‐repeat as a main factor in exhibiting these for better and for worse characteristics. However, human studies lack the genetic and environmental control offered by animal experiments, complicating assessment of causal relations. To study differential susceptibility in an animal model, we exposed Drd4+/− mice and control litter mates to a limited nesting/bedding (LN), standard nesting (SN) or communal nesting (CN) rearing environment from postnatal day (P) 2‐14. Puberty onset was examined from P24 to P36 and adult females were assessed on maternal care towards their own offspring. In both males and females, LN reared mice showed a delay in puberty onset that was partly mediated by a reduction in body weight at weaning, irrespective of Drd4 genotype. During adulthood, LN reared females exhibited characteristics of poor maternal care, whereas dams reared in CN environments showed lower rates of unpredictability towards their own offspring. Differential susceptibility was observed only for licking/grooming levels of female offspring towards their litter; LN reared Drd4+/− mice exhibited the lowest and CN reared Drd4+/− mice the highest levels of licking/grooming. These results indicate that both genetic and early‐environmental factors play an important role in shaping maternal care of the offspring for better and for worse.
Collapse
Affiliation(s)
- Jelle Knop
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Faculty of Social and Behavioural Sciences, Leiden University, Leiden, The Netherlands
| | - Marinus H van IJzendoorn
- Department of Psychology, Education and Child Studies, Erasmus University Rotterdam, Rotterdam, The Netherlands.,Primary Care Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | | | - Marian Joëls
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rixt van der Veen
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Faculty of Social and Behavioural Sciences, Leiden University, Leiden, The Netherlands
| |
Collapse
|
26
|
Köfalvi A, Moreno E, Cordomí A, Cai NS, Fernández-Dueñas V, Ferreira SG, Guixà-González R, Sánchez-Soto M, Yano H, Casadó-Anguera V, Cunha RA, Sebastião AM, Ciruela F, Pardo L, Casadó V, Ferré S. Control of glutamate release by complexes of adenosine and cannabinoid receptors. BMC Biol 2020; 18:9. [PMID: 31973708 PMCID: PMC6979073 DOI: 10.1186/s12915-020-0739-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/13/2020] [Indexed: 12/20/2022] Open
Abstract
Background It has been hypothesized that heteromers of adenosine A2A receptors (A2AR) and cannabinoid CB1 receptors (CB1R) localized in glutamatergic nerve terminals mediate the integration of adenosine and endocannabinoid signaling involved in the modulation of striatal excitatory neurotransmission. Previous studies have demonstrated the existence of A2AR-CB1R heteromers in artificial cell systems. A dependence of A2AR signaling for the Gi protein-mediated CB1R signaling was described as one of its main biochemical characteristics. However, recent studies have questioned the localization of functionally significant A2AR-CB1R heteromers in striatal glutamatergic terminals. Results Using a peptide-interfering approach combined with biophysical and biochemical techniques in mammalian transfected cells and computational modeling, we could establish a tetrameric quaternary structure of the A2AR-CB1R heterotetramer. This quaternary structure was different to the also tetrameric structure of heteromers of A2AR with adenosine A1 receptors or dopamine D2 receptors, with different heteromeric or homomeric interfaces. The specific quaternary structure of the A2A-CB1R, which depended on intermolecular interactions involving the long C-terminus of the A2AR, determined a significant A2AR and Gs protein-mediated constitutive activation of adenylyl cyclase. Using heteromer-interfering peptides in experiments with striatal glutamatergic terminals, we could then demonstrate the presence of functionally significant A2AR-CB1R heteromers with the same biochemical characteristics of those studied in mammalian transfected cells. First, either an A2AR agonist or an A2AR antagonist allosterically counteracted Gi-mediated CB1R agonist-induced inhibition of depolarization-induced glutamate release. Second, co-application of both an A2AR agonist and an antagonist cancelled each other effects. Finally, a CB1R agonist inhibited glutamate release dependent on a constitutive activation of A2AR by a canonical Gs-Gi antagonistic interaction at the adenylyl cyclase level. Conclusions We demonstrate that the well-established cannabinoid-induced inhibition of striatal glutamate release can mostly be explained by a CB1R-mediated counteraction of the A2AR-mediated constitutive activation of adenylyl cyclase in the A2AR-CB1R heteromer.
Collapse
Affiliation(s)
- Attila Köfalvi
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Estefanía Moreno
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, and Institute of Biomedicine, University of Barcelona, 08028, Barcelona, Spain
| | - Arnau Cordomí
- Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Ning-Sheng Cai
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Victor Fernández-Dueñas
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Samira G Ferreira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Ramón Guixà-González
- Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain
| | - Marta Sánchez-Soto
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Hideaki Yano
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Verònica Casadó-Anguera
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, and Institute of Biomedicine, University of Barcelona, 08028, Barcelona, Spain
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana Maria Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Francisco Ciruela
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain. .,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.
| | - Leonardo Pardo
- Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain.
| | - Vicent Casadó
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, and Institute of Biomedicine, University of Barcelona, 08028, Barcelona, Spain.
| | - Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
27
|
Rauschenberger L, Knorr S, Al-Zuraiqi Y, Tovote P, Volkmann J, Ip CW. Striatal dopaminergic dysregulation and dystonia-like movements induced by sensorimotor stress in a pharmacological mouse model of rapid-onset dystonia-parkinsonism. Exp Neurol 2020; 323:113109. [DOI: 10.1016/j.expneurol.2019.113109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/18/2019] [Accepted: 11/07/2019] [Indexed: 12/21/2022]
|
28
|
Cromwell HC. Translating striatal activity from brain slice to whole animal neurophysiology: A guide for neuroscience research integrating diverse levels of analysis. J Neurosci Res 2019; 97:1528-1545. [PMID: 31257656 DOI: 10.1002/jnr.24480] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/20/2019] [Accepted: 05/22/2019] [Indexed: 01/09/2023]
Abstract
An important goal of this review is highlighting research in neuroscience as examples of multilevel functional and anatomical analyses addressing basic science issues and applying results to the understanding of diverse disorders. The research of Dr. Michael Levine, a leader in neuroscience, exemplifies this approach by uncovering fundamental properties of basal ganglia function and translating these findings to clinical applications. The review focuses on neurophysiological research connecting results from in vitro and in vivo recordings. A second goal is to utilize these research connections to produce novel, accurate descriptions for corticostriatal processing involved in varied, complex functions. Medium spiny neurons in striatum act as integrators combining input with baseline activity creating motivational "events." Basic research on corticostriatal synapses is described and links developed to issues with clinical relevance such as inhibitory gating, self-injurious behavior, and relative reward valuation. Work is highlighted on dopamine-glutamate interactions. Individual medium spiny neurons express both D1 and D2 receptors and encode information in a bivalent manner depending upon the mix of receptors involved. Current work on neurophysiology of reward processing has taken advantage of these basic approaches at the cellular and molecular levels. Future directions in studying physiology of reward processing and action sequencing could profit by incorporating the divergent ways dopamine modulates incoming neurochemical signals. Primary investigators leading research teams should mirror Mike Levine's efforts in "climbing the mountain" of scientific inquiry by performing analyses at different levels of inquiry, integrating the findings, and building comprehensive answers to problems unsolvable without this bold approach.
Collapse
Affiliation(s)
- Howard Casey Cromwell
- Department of Psychology and John Paul Scott Center for Neuroscience, Mind and Behavior, Bowling Green State University, Bowling Green, Ohio
| |
Collapse
|
29
|
D3 and D1 receptors: The Yin and Yang in the treatment of restless legs syndrome with dopaminergics. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2019; 84:79-100. [PMID: 31229178 DOI: 10.1016/bs.apha.2019.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Dopaminergic treatments targeting the D3 receptor subtype to reduce the symptoms of RLS show substantial initial clinical benefits but fail to maintain their efficacy over time. Sensorimotor circuits in the spinal cord are the gateway for the sensory processing of the symptoms and critical for the associated leg movements that relieve the symptoms and the periodic limb movements that often develop during sleep. There is a high preponderance of the inhibitory D3 receptor in the sensory-processing areas of the spinal cord (dorsal horn), whereas the motor areas in the ventral horn more strongly express the excitatory D1 receptor subtype. D3 and D1 receptors can form functional heteromeric ensembles that influence each other. In the spinal cord, long-term treatment with D3 receptor agonists is associated with the upregulation of the D1 receptor subtype and block of D1 receptor function at this stage can restore the D3 receptor effect. Alternate scenarios for a role of dopamine involve a role for the D5 receptor in regulating motor excitability and for the D4 receptor subtype in controlling D3-like effects. A model emerges that proposes that the behavioral changes in RLS, while responsive to D3 receptor agonists, may be ultimately be the result of unmasked increased D1-like receptor activities.
Collapse
|
30
|
Kuo HY, Liu FC. Synaptic Wiring of Corticostriatal Circuits in Basal Ganglia: Insights into the Pathogenesis of Neuropsychiatric Disorders. eNeuro 2019; 6:ENEURO.0076-19.2019. [PMID: 31097624 PMCID: PMC6553570 DOI: 10.1523/eneuro.0076-19.2019] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 04/26/2019] [Accepted: 05/07/2019] [Indexed: 02/06/2023] Open
Abstract
The striatum is a key hub in the basal ganglia for processing neural information from the sensory, motor, and limbic cortices. The massive and diverse cortical inputs entering the striatum allow the basal ganglia to perform a repertoire of neurological functions ranging from basic level of motor control to high level of cognition. The heterogeneity of the corticostriatal circuits, however, also renders the system susceptible to a repertoire of neurological diseases. Clinical and animal model studies have indicated that defective development of the corticostriatal circuits is linked to various neuropsychiatric disorders, including attention-deficit hyperactivity disorder (ADHD), Tourette syndrome, obsessive-compulsive disorder (OCD), autism spectrum disorder (ASD), and schizophrenia. Importantly, many neuropsychiatric disease-risk genes have been found to form the molecular building blocks of the circuit wiring at the synaptic level. It is therefore imperative to understand how corticostriatal connectivity is established during development. Here, we review the construction during development of these corticostriatal circuits at the synaptic level, which should provide important insights into the pathogenesis of neuropsychiatric disorders related to the basal ganglia and help the development of appropriate therapies for these diseases.
Collapse
Affiliation(s)
- Hsiao-Ying Kuo
- Institute of Neuroscience, National Yang-Ming University, Taipei 11221, Taiwan
| | - Fu-Chin Liu
- Institute of Neuroscience, National Yang-Ming University, Taipei 11221, Taiwan
- Brain Research Center, National Yang-Ming University, Taipei 11221, Taiwan
| |
Collapse
|
31
|
Cui J, Fu Y, Lu R, Bi Y, Zhang L, Zhang C, Aschner M, Li X, Chen R. Metabolomics analysis explores the rescue to neurobehavioral disorder induced by maternal PM 2.5 exposure in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 169:687-695. [PMID: 30500738 DOI: 10.1016/j.ecoenv.2018.11.037] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 10/17/2018] [Accepted: 11/09/2018] [Indexed: 06/09/2023]
Abstract
Reproductive epidemiological studies have suggested associations between perinatal exposure to fine particulate matter (PM2.5) and adverse birth outcomes. To explore the effects of early prenatal exposure to PM2.5 on subsequent generations, pregnant mice were exposed to PM2.5 or filtered clean air in whole body dynamic exposure chambers for 14 consecutive days from gestation day (GD) 1.5 to GD15.5. Neurobehavioral tests showed that spontaneous locomotion and exploratory behaviors in the offspring were significantly enhanced in the open field test. Meanwhile, metabolomics analysis suggested activation of dopamine pathway while inhibition of glycine pathway in murine brains. Administration of the DRD4 antagonist, clozapine; or supplementation of glycine receptor agonist, taurine, to mice offspring attenuated the locomotor hyperactivities to levels indistinguishable from controls. These data provide strong evidence that maternal exposure to air pollution might increase the risk for neural disorders in the offspring during critical periods of brain development.
Collapse
Affiliation(s)
- Jian Cui
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education School of Public Health, Southeast University, Nanjing 210009, China
| | - You Fu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education School of Public Health, Southeast University, Nanjing 210009, China; School of Continuing Education, Nanjing Medical University, Nanjing 211166, China
| | - Runze Lu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education School of Public Health, Southeast University, Nanjing 210009, China
| | - Yuan Bi
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education School of Public Health, Southeast University, Nanjing 210009, China
| | - Li Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education School of Public Health, Southeast University, Nanjing 210009, China
| | - Chengcheng Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education School of Public Health, Southeast University, Nanjing 210009, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer 209, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Xiaobo Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education School of Public Health, Southeast University, Nanjing 210009, China.
| | - Rui Chen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education School of Public Health, Southeast University, Nanjing 210009, China; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
32
|
Reduced response to gabapentin enacarbil in restless legs syndrome following long-term dopaminergic treatment. Sleep Med 2019; 55:74-80. [DOI: 10.1016/j.sleep.2018.11.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/04/2018] [Accepted: 11/26/2018] [Indexed: 11/20/2022]
|
33
|
Cortés A, Casadó-Anguera V, Moreno E, Casadó V. The heterotetrameric structure of the adenosine A 1-dopamine D 1 receptor complex: Pharmacological implication for restless legs syndrome. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2019; 84:37-78. [PMID: 31229177 DOI: 10.1016/bs.apha.2019.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Dopaminergic and purinergic signaling play a pivotal role in neurological diseases associated with motor symptoms, including Parkinson's disease (PD), multiple sclerosis, amyotrophic lateral sclerosis, Huntington disease, Restless Legs Syndrome (RLS), spinal cord injury (SCI), and ataxias. Extracellular dopamine and adenosine exert their functions interacting with specific dopamine (DR) or adenosine (AR) receptors, respectively, expressed on the surface of target cells. These receptors are members of the family A of G protein-coupled receptors (GPCRs), which is the largest protein superfamily in mammalian genomes. GPCRs are target of about 40% of all current marketed drugs, highlighting their importance in clinical medicine. The striatum receives the densest dopamine innervations and contains the highest density of dopamine receptors. The modulatory role of adenosine on dopaminergic transmission depends largely on the existence of antagonistic interactions mediated by specific subtypes of DRs and ARs, the so-called A2AR-D2R and A1R-D1R interactions. Due to the dopamine/adenosine antagonism in the CNS, it was proposed that ARs and DRs could form heteromers in the neuronal cell surface. Therefore, adenosine can affect dopaminergic signaling through receptor-receptor interactions and by modulations in their shared intracellular pathways in the striatum and spinal cord. In this work we describe the allosteric modulations between GPCR protomers, focusing in those of adenosine and dopamine within the A1R-D1R heteromeric complex, which is involved in RLS. We also propose that the knowledge about the intricate allosteric interactions within the A1R-D1R heterotetramer, may facilitate the treatment of motor alterations, not only when the dopamine pathway is hyperactivated (RLS, chorea, etc.) but also when motor function is decreased (SCI, aging, PD, etc.).
Collapse
Affiliation(s)
- Antoni Cortés
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Verònica Casadó-Anguera
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Estefanía Moreno
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Vicent Casadó
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
34
|
Xu L, Hu Y, Huang L, Liu Y, Wang B, Xie L, Hu Z. The association between attention deficit hyperactivity disorder and general anaesthesia - a narrative review. Anaesthesia 2018; 74:57-63. [PMID: 30511754 DOI: 10.1111/anae.14496] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2018] [Indexed: 11/30/2022]
Affiliation(s)
- L. Xu
- Department of Anesthesiology; Women's Hospital; Zhejiang University School of Medicine; Hangzhou China
| | - Y. Hu
- Department of Cell Biology; Yale University; New Haven CT USA
| | - L. Huang
- Department of Anesthesiology; The Children's Hospital; Zhejiang University School of Medicine; Hangzhou China
| | - Y. Liu
- Department of Anesthesiology; The Children's Hospital; Zhejiang University School of Medicine; Hangzhou China
| | - B. Wang
- Department of Anesthesiology; The Children's Hospital; Zhejiang University School of Medicine; Hangzhou China
| | - L. Xie
- Department of Anesthesiology; The Children's Hospital; Zhejiang University School of Medicine; Hangzhou China
| | - Z. Hu
- Department of Anesthesiology; The Children's Hospital; Zhejiang University School of Medicine; Hangzhou China
| |
Collapse
|
35
|
Sánchez-Soto M, Yano H, Cai NS, Casadó-Anguera V, Moreno E, Casadó V, Ferré S. Revisiting the Functional Role of Dopamine D 4 Receptor Gene Polymorphisms: Heteromerization-Dependent Gain of Function of the D 4.7 Receptor Variant. Mol Neurobiol 2018; 56:4778-4785. [PMID: 30387076 DOI: 10.1007/s12035-018-1413-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 10/24/2018] [Indexed: 12/28/2022]
Abstract
The two most common polymorphisms of the human DRD4 gene encode a dopamine D4 receptor (D4R) with four or seven repeats of a proline-rich sequence of 16 amino acids (D4.4R or D4.7R). Although the seven-repeat polymorphism has been repeatedly associated with attention-deficit hyperactivity disorder and substance use disorders, the differential functional properties between D4.4R and D4.7R remained enigmatic until recent electrophysiological and optogenetic-microdialysis experiments indicated a gain of function of D4.7R. Since no clear differences in the biochemical properties of individual D4.4R and D4.7R have been reported, it was previously suggested that those differences emerge upon heteromerization with dopamine D2 receptor (D2R), which co-localizes with D4R in the brain. However, contrary to a gain of function, experiments in mammalian transfected cells suggested that heteromerization with D2R results in lower MAPK signaling by D4.7R as compared to D4.4R. In the present study, we readdressed the question of functional differences of D4.4R and D4.7R forming homomers or heteromers with the short isoform of D2R (D2SR), using a functional bioluminescence resonance energy transfer (BRET) assay that allows the measurement of ligand-induced changes in the interaction between G protein-coupled receptors (GPCRs) forming homomers or heteromers with their cognate G protein. Significant functional and pharmacological differences between D4.4R and D4.7R were only evident upon heteromerization with the short isoform of D2R (D2SR). The most dramatic finding was a significant increase and decrease in the constitutive activity of D2S upon heteromerization with D4.7R and D4.4R, respectively, providing the first clear mechanism for a functional difference between both products of polymorphic variants and for a gain of function of the D4.7R.
Collapse
Affiliation(s)
- Marta Sánchez-Soto
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Hideaki Yano
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Ning-Sheng Cai
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Verònica Casadó-Anguera
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona, University of Barcelona, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08028, Barcelona, Spain
| | - Estefanía Moreno
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona, University of Barcelona, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08028, Barcelona, Spain
| | - Vicent Casadó
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona, University of Barcelona, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08028, Barcelona, Spain
| | - Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
36
|
Elam KK, DiLalla LF. Minute-to-minute trajectories of child unresponsiveness and parent sensitivity in parent-child interactions: The role of DRD4. SOCIAL DEVELOPMENT 2018. [DOI: 10.1111/sode.12308] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Kit K. Elam
- T. Denny Sanford School of Social and Family Dynamics; Arizona State University; Tempe Arizona
| | - Lisabeth F. DiLalla
- School of Medicine; Department of Family and Community Medicine; Southern Illinois University; Carbondale Illinois
| |
Collapse
|
37
|
Ferré S, Bonaventura J, Zhu W, Hatcher-Solis C, Taura J, Quiroz C, Cai NS, Moreno E, Casadó-Anguera V, Kravitz AV, Thompson KR, Tomasi DG, Navarro G, Cordomí A, Pardo L, Lluís C, Dessauer CW, Volkow ND, Casadó V, Ciruela F, Logothetis DE, Zwilling D. Essential Control of the Function of the Striatopallidal Neuron by Pre-coupled Complexes of Adenosine A 2A-Dopamine D 2 Receptor Heterotetramers and Adenylyl Cyclase. Front Pharmacol 2018; 9:243. [PMID: 29686613 PMCID: PMC5900444 DOI: 10.3389/fphar.2018.00243] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 03/05/2018] [Indexed: 01/10/2023] Open
Abstract
The central adenosine system and adenosine receptors play a fundamental role in the modulation of dopaminergic neurotransmission. This is mostly achieved by the strategic co-localization of different adenosine and dopamine receptor subtypes in the two populations of striatal efferent neurons, striatonigral and striatopallidal, that give rise to the direct and indirect striatal efferent pathways, respectively. With optogenetic techniques it has been possible to dissect a differential role of the direct and indirect pathways in mediating "Go" responses upon exposure to reward-related stimuli and "NoGo" responses upon exposure to non-rewarded or aversive-related stimuli, respectively, which depends on their different connecting output structures and their differential expression of dopamine and adenosine receptor subtypes. The striatopallidal neuron selectively expresses dopamine D2 receptors (D2R) and adenosine A2A receptors (A2AR), and numerous experiments using multiple genetic and pharmacological in vitro, in situ and in vivo approaches, demonstrate they can form A2AR-D2R heteromers. It was initially assumed that different pharmacological interactions between dopamine and adenosine receptor ligands indicated the existence of different subpopulations of A2AR and D2R in the striatopallidal neuron. However, as elaborated in the present essay, most evidence now indicates that all interactions can be explained with a predominant population of striatal A2AR-D2R heteromers forming complexes with adenylyl cyclase subtype 5 (AC5). The A2AR-D2R heteromer has a tetrameric structure, with two homodimers, which allows not only multiple allosteric interactions between different orthosteric ligands, agonists, and antagonists, but also the canonical Gs-Gi antagonistic interaction at the level of AC5. We present a model of the function of the A2AR-D2R heterotetramer-AC5 complex, which acts as an integrative device of adenosine and dopamine signals that determine the excitability and gene expression of the striatopallidal neurons. The model can explain most behavioral effects of A2AR and D2R ligands, including the psychostimulant effects of caffeine. The model is also discussed in the context of different functional striatal compartments, mainly the dorsal and the ventral striatum. The current accumulated knowledge of the biochemical properties of the A2AR-D2R heterotetramer-AC5 complex offers new therapeutic possibilities for Parkinson's disease, schizophrenia, SUD and other neuropsychiatric disorders with dysfunction of dorsal or ventral striatopallidal neurons.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Jordi Bonaventura
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Wendy Zhu
- Circuit Therapeutics, Inc., Menlo Park, CA, United States
| | - Candice Hatcher-Solis
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Jaume Taura
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina i Ciències de la Salut, IDIBELL, Universitat de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - César Quiroz
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Ning-Sheng Cai
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Estefanía Moreno
- Center for Biomedical Research in Neurodegenerative Diseases Network, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona, University of Barcelona, Barcelona, Spain
| | - Verónica Casadó-Anguera
- Center for Biomedical Research in Neurodegenerative Diseases Network, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona, University of Barcelona, Barcelona, Spain
| | - Alexxai V Kravitz
- Eating and Addiction Section, Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Intramural Research Program, National Institutes of Health, Bethesda, MD, United States
| | | | - Dardo G Tomasi
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, National Institutes of Health, Rockville, MD, United States
| | - Gemma Navarro
- Department of Biochemistry and Physiology, Faculty of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Arnau Cordomí
- Laboratory of Computational Medicine, School of Medicine, Autonomous University of Barcelona, Bellaterra, Spain
| | - Leonardo Pardo
- Laboratory of Computational Medicine, School of Medicine, Autonomous University of Barcelona, Bellaterra, Spain
| | - Carme Lluís
- Center for Biomedical Research in Neurodegenerative Diseases Network, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona, University of Barcelona, Barcelona, Spain
| | - Carmen W Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Nora D Volkow
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Intramural Research Program, National Institutes of Health, Rockville, MD, United States
| | - Vicent Casadó
- Center for Biomedical Research in Neurodegenerative Diseases Network, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona, University of Barcelona, Barcelona, Spain
| | - Francisco Ciruela
- Unitat de Farmacologia, Departament de Patologia i Terapèutica Experimental, Facultat de Medicina i Ciències de la Salut, IDIBELL, Universitat de Barcelona, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Diomedes E Logothetis
- Department of Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, United States
| | | |
Collapse
|
38
|
Gioia DA, Xu M, Wayman WN, Woodward JJ. Effects of drugs of abuse on channelrhodopsin-2 function. Neuropharmacology 2018; 135:316-327. [PMID: 29580953 DOI: 10.1016/j.neuropharm.2018.03.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 01/16/2023]
Abstract
Channelrhodopsins are light activated ion channels used extensively over the past decade to probe the function of genetically defined neuronal populations and distinct neural circuits with high temporal and spatial precision. The widely used Channelrhodopsin-2 variant (ChR2) is an excitatory opsin that undergoes conformational changes in response to blue light, allowing non-selective passage of protons and cations across the plasma membrane thus leading to depolarization. In the addiction neuroscience field, opsins such as ChR2 provide a means to disambiguate the overlapping circuitry involved in mediating the reinforcing and aversive effects of drugs of abuse as well as to determine the plasticity that can occur in these circuits during the development of dependence. Although ChR2 has been widely used in animal models of drug and alcohol self-administration, direct effects of drugs of abuse on ChR2 function may confound its use and lead to misinterpretation of data. As a variety of neuronal ion channels are primary targets of various drugs of abuse, it is critical to determine whether ChR2-mediated currents are modulated by these drugs. In this study, we performed whole-cell electrophysiological recordings in HEK293 cells expressing the commonly used ChR2(H134R) variant and examined the effects of various drugs of abuse and other commonly used agents on light-induced currents. We found no differences in ChR2-mediated currents in the presence of 30 μM nicotine, 30 μM cocaine, 100 μM methamphetamine or 3 mM toluene. Similarly, ChR2 currents were insensitive to 30 mM ethanol but higher concentrations (100-300 mM) produced significant effects on the desensitization and amplitude of light-evoked currents. Tetrahydrocannabinol (1-10 μM) and morphine (30-100 μM) significantly inhibited ChR2 currents while the cannabinoid receptor antagonist AM-251 had no effect. The sodium channel blocker tetrodotoxin (5 μM) and the generic channel blocker/contrast agent gadolinium chloride (10 mM) also reduced ChR2 currents while the divalent ion magnesium (10 mM) had no effect. Together, the results from this study highlight the importance of conducting appropriate control experiments when testing new compounds in combination with optogenetic approaches.
Collapse
Affiliation(s)
- Dominic A Gioia
- Medical University of South Carolina, Institute of Psychiatry, 67 President St, Charleston, SC, 29425, United States.
| | - Minfu Xu
- Medical University of South Carolina, Institute of Psychiatry, 67 President St, Charleston, SC, 29425, United States.
| | - Wesley N Wayman
- Medical University of South Carolina, Institute of Psychiatry, 67 President St, Charleston, SC, 29425, United States.
| | - John J Woodward
- Medical University of South Carolina, Institute of Psychiatry, 67 President St, Charleston, SC, 29425, United States.
| |
Collapse
|
39
|
Ferré S, Quiroz C, Guitart X, Rea W, Seyedian A, Moreno E, Casadó-Anguera V, Díaz-Ríos M, Casadó V, Clemens S, Allen RP, Earley CJ, García-Borreguero D. Pivotal Role of Adenosine Neurotransmission in Restless Legs Syndrome. Front Neurosci 2018; 11:722. [PMID: 29358902 PMCID: PMC5766678 DOI: 10.3389/fnins.2017.00722] [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: 10/30/2017] [Accepted: 12/11/2017] [Indexed: 11/13/2022] Open
Abstract
The symptomatology of Restless Legs Syndrome (RLS) includes periodic leg movements during sleep (PLMS), dysesthesias, and hyperarousal. Alterations in the dopaminergic system, a presynaptic hyperdopaminergic state, seem to be involved in PLMS, while alterations in glutamatergic neurotransmission, a presynaptic hyperglutamatergic state, seem to be involved in hyperarousal and also PLMS. Brain iron deficiency (BID) is well-recognized as a main initial pathophysiological mechanism of RLS. BID in rodents have provided a pathogenetic model of RLS that recapitulates the biochemical alterations of the dopaminergic system of RLS, although without PLMS-like motor abnormalities. On the other hand, BID in rodents reproduces the circadian sleep architecture of RLS, indicating the model could provide clues for the hyperglutamatergic state in RLS. We recently showed that BID in rodents is associated with changes in adenosinergic transmission, with downregulation of adenosine A1 receptors (A1R) as the most sensitive biochemical finding. It was hypothesized that A1R downregulation leads to hypersensitive striatal glutamatergic terminals and facilitation of striatal dopamine release. Hypersensitivity of striatal glutamatergic terminals was demonstrated by an optogenetic-microdialysis approach in the rodent with BID, indicating that it could represent a main pathogenetic factor that leads to PLMS in RLS. In fact, the dopaminergic agonists pramipexole and ropinirole and the α2δ ligand gabapentin, used in the initial symptomatic treatment of RLS, completely counteracted optogenetically-induced glutamate release from both normal and BID-induced hypersensitive corticostriatal glutamatergic terminals. It is a main tenet of this essay that, in RLS, a single alteration in the adenosinergic system, downregulation of A1R, disrupts the adenosine-dopamine-glutamate balance uniquely controlled by adenosine and dopamine receptor heteromers in the striatum and also the A1R-mediated inhibitory control of glutamatergic neurotransmission in the cortex and other non-striatal brain areas, which altogether determine both PLMS and hyperarousal. Since A1R agonists would be associated with severe cardiovascular effects, it was hypothesized that inhibitors of nucleoside equilibrative transporters, such as dipyridamole, by increasing the tonic A1R activation mediated by endogenous adenosine, could represent a new alternative therapeutic strategy for RLS. In fact, preliminary clinical data indicate that dipyridamole can significantly improve the symptomatology of RLS.
Collapse
Affiliation(s)
- Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - César Quiroz
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Xavier Guitart
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - William Rea
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Arta Seyedian
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Estefanía Moreno
- Center for Biomedical Research in Neurodegenerative Diseases Network and Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona, University of Barcelona, Barcelona, Spain
| | - Verònica Casadó-Anguera
- Center for Biomedical Research in Neurodegenerative Diseases Network and Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona, University of Barcelona, Barcelona, Spain
| | - Manuel Díaz-Ríos
- Department of Anatomy and Neurobiology and Institute of Neurobiology, University of Puerto Rico, San Juan, PR, United States
| | - Vicent Casadó
- Center for Biomedical Research in Neurodegenerative Diseases Network and Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Institute of Biomedicine of the University of Barcelona, University of Barcelona, Barcelona, Spain
| | - Stefan Clemens
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Richard P Allen
- Center for Restless Legs Study, Department of Neurology, Johns Hopkins University, Baltimore, MD, United States
| | - Christopher J Earley
- Center for Restless Legs Study, Department of Neurology, Johns Hopkins University, Baltimore, MD, United States
| | | |
Collapse
|
40
|
Yepes G, Guitart X, Rea W, Newman AH, Allen RP, Earley CJ, Quiroz C, Ferré S. Targeting hypersensitive corticostriatal terminals in restless legs syndrome. Ann Neurol 2017; 82:951-960. [PMID: 29171915 DOI: 10.1002/ana.25104] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 11/16/2017] [Accepted: 11/16/2017] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The first aim was to demonstrate a previously hypothesized increased sensitivity of corticostriatal glutamatergic terminals in the rodent with brain iron deficiency (BID), a pathogenetic model of restless legs syndrome (RLS). The second aim was to determine whether these putative hypersensitive terminals could constitute a significant target for drugs effective in RLS, including dopamine agonists (pramipexole and ropinirole) and α2 δ ligands (gabapentin). METHODS A recently introduced in vivo optogenetic-microdialysis approach was used, which allows the measurement of the extracellular concentration of glutamate upon local light-induced stimulation of corticostriatal glutamatergic terminals. The method also allows analysis of the effect of local perfusion of compounds within the same area being sampled for glutamate. RESULTS BID rats showed hypersensitivity of corticostriatal glutamatergic terminals (lower frequency of optogenetic stimulation to induce glutamate release). Both hypersensitive and control glutamatergic terminals were significant targets for locally perfused pramipexole, ropinirole, and gabapentin, which significantly counteracted optogenetically induced glutamate release. The use of selective antagonists demonstrated the involvement of dopamine D4 and D2 receptor subtypes in the effects of pramipexole. INTERPRETATION Hypersensitivity of corticostriatal glutamatergic terminals can constitute a main pathogenetic mechanism of RLS symptoms. Selective D4 receptor agonists, by specifically targeting these terminals, should provide a new efficient treatment with fewer secondary effects. Ann Neurol 2017;82:951-960.
Collapse
Affiliation(s)
- Gabriel Yepes
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD
| | - Xavier Guitart
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD
| | - William Rea
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD
| | - Amy H Newman
- Medicinal Chemistry Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD
| | - Richard P Allen
- Center for Restless Legs Study, Department of Neurology, Johns Hopkins University, Baltimore, MD
| | - Christopher J Earley
- Center for Restless Legs Study, Department of Neurology, Johns Hopkins University, Baltimore, MD
| | - César Quiroz
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD
| | - Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD
| |
Collapse
|
41
|
Fernández-Dueñas V, Pérez-Arévalo A, Altafaj X, Ferré S, Ciruela F. Adenosine A 1-A 2A Receptor Heteromer as a Possible Target for Early-Onset Parkinson's Disease. Front Neurosci 2017; 11:652. [PMID: 29213228 PMCID: PMC5702635 DOI: 10.3389/fnins.2017.00652] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 11/09/2017] [Indexed: 11/13/2022] Open
Affiliation(s)
- Víctor Fernández-Dueñas
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Andrea Pérez-Arévalo
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Xavier Altafaj
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Sergi Ferré
- Integrative Neurobiology Section, National Institute on Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, United States
| | - Francisco Ciruela
- Unitat de Farmacologia, Departament Patologia i Terapèutica Experimental, Facultat de Medicina, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
42
|
Lindsley CW, Hopkins CR. Return of D4 Dopamine Receptor Antagonists in Drug Discovery. J Med Chem 2017; 60:7233-7243. [DOI: 10.1021/acs.jmedchem.7b00151] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
| | - Corey R. Hopkins
- Department
of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198-6125, United States
| |
Collapse
|