1
|
Brown KA, Stramiello M, Clark JK, Wagner JJ. Postsynaptic dopamine D 3 receptors selectively modulate μ-opioid receptor-expressing GABAergic inputs onto CA1 pyramidal cells in the rat ventral hippocampus. J Neurophysiol 2024; 132:2002-2011. [PMID: 39570291 PMCID: PMC11968057 DOI: 10.1152/jn.00353.2024] [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: 08/08/2024] [Revised: 10/07/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024] Open
Abstract
Although the actions of dopamine throughout the brain are clearly linked to motivation and cognition, the specific role(s) of dopamine in the CA1 subfield of the ventral hippocampus (vH) is unresolved. Prior preclinical studies suggest that dopamine D3 receptors (D3Rs) expressed on CA1 pyramidal cells exhibit a unique capacity to modulate mechanisms of long-term synaptic plasticity, but less is known about how interneuronal inputs modulate these cells. We hypothesized that inputs from μ-opioid receptor (MOR)-expressing inhibitory interneurons selectively modulate the activity of postsynaptic D3Rs expressed on CA1 principal cells to shape neurotransmission in the rat vH. We used the whole cell voltage-clamp technique to test this hypothesis by measuring evoked inhibitory postsynaptic currents (eIPSCs) from CA1 principal cells in vH slices or GABAA currents from acutely dissociated vH neurons. The eIPSC response recorded from CA1 neurons in vH slices was inhibited by either the MOR agonist DAMGO or the D3R agonist PD128907, but pretreatment with DAMGO occluded any further inhibition by PD128907. GABAA currents measured in acutely dissociated vH CA1 neurons were inhibited by D3R activation via PD128907, consistent with postsynaptic localization of D3 receptors. Kinetic alterations induced by the neuromodulatory agonists are consistent with selective targeting of postsynaptic D3Rs expressed on CA1 principal cells by MOR-expressing GABAergic inputs. Our findings suggest that postsynaptic D3R-mediated modulation of MOR-expressing GABAergic inputs is a site at which dopaminergic and opioidergic activity may contribute to disinhibition of vH excitatory neurotransmission and, thus, influence critical physiological processes such as synaptic plasticity and network oscillations.NEW & NOTEWORTHY We report that the activity of an inhibitory synapse on CA1 pyramidal cells in the rat ventral hippocampus is shaped by heterogeneous neuromodulators. Specifically, postsynaptic dopamine D3 receptors on ventral hippocampal CA1 pyramidal neurons are selectively targeted by an inhibitory input from µ-opioid receptor-expressing GABAergic terminals.
Collapse
Affiliation(s)
- Kyle A Brown
- Department of Physiology & Pharmacology, University of Georgia, Athens, Georgia, United States
- Interdisciplinary Toxicology Program, University of Georgia, Athens, Georgia, United States
| | - Michael Stramiello
- Department of Physiology & Pharmacology, University of Georgia, Athens, Georgia, United States
- Neuroscience PhD Program, University of Georgia, Athens, Georgia, United States
| | - Jason K Clark
- Department of Physiology & Pharmacology, University of Georgia, Athens, Georgia, United States
| | - John J Wagner
- Department of Physiology & Pharmacology, University of Georgia, Athens, Georgia, United States
- Interdisciplinary Toxicology Program, University of Georgia, Athens, Georgia, United States
- Neuroscience PhD Program, University of Georgia, Athens, Georgia, United States
| |
Collapse
|
2
|
Shen H, Ma Z, Hans E, Duan Y, Bi GH, Chae YC, Bonifazi A, Battiti FO, Newman AH, Xi ZX, Yang Y. Involvement of dopamine D3 receptor in impulsive choice decision-making in male rats. Neuropharmacology 2024; 257:110051. [PMID: 38917939 PMCID: PMC11401648 DOI: 10.1016/j.neuropharm.2024.110051] [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: 04/13/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 06/27/2024]
Abstract
Impulsive decision-making has been linked to impulse control disorders and substance use disorders. However, the neural mechanisms underlying impulsive choice are not fully understood. While previous PET imaging and autoradiography studies have shown involvement of dopamine and D2/3 receptors in impulsive behavior, the roles of distinct D1, D2, and D3 receptors in impulsive decision-making remain unclear. In this study, we used a food reward delay-discounting task (DDT) to identify low- and high-impulsive rats, in which low-impulsive rats exhibited preference for large delayed reward over small immediate rewards, while high-impulsive rats showed the opposite preference. We then examined D1, D2, and D3 receptor gene expression using RNAscope in situ hybridization assays. We found that high-impulsive male rats exhibited lower levels of D2 and D3, and particularly D3, receptor expression in the nucleus accumbens (NAc), with no significant changes in the insular, prelimbic, and infralimbic cortices. Based on these findings, we further explored the role of the D3 receptor in impulsive decision-making. Systemic administration of a selective D3 receptor agonist (FOB02-04) significantly reduced impulsive choices in high-impulsive rats but had no effects in low-impulsive rats. Conversely, a selective D3 receptor antagonist (VK4-116) produced increased both impulsive and omission choices in both groups of rats. These findings suggest that impulsive decision-making is associated with a reduction in D3 receptor expression in the NAc. Selective D3 receptor agonists, but not antagonists, may hold therapeutic potentials for mitigating impulsivity in high-impulsive subjects.
Collapse
Affiliation(s)
- Hui Shen
- Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Zilu Ma
- Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Emma Hans
- Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Ying Duan
- Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Guo-Hua Bi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Yurim C Chae
- Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Alessandro Bonifazi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Francisco O Battiti
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Amy Hauck Newman
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Zheng-Xiong Xi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA.
| | - Yihong Yang
- Neuroimaging Research Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA.
| |
Collapse
|
3
|
Lei J, Tang LL, Jing R, You HJ. Antinociceptive role of the thalamic dopamine D3 receptor in descending modulation of intramuscular formalin-induced muscle nociception in a rat model of Parkinson's disease. Exp Neurol 2024; 379:114846. [PMID: 38879111 DOI: 10.1016/j.expneurol.2024.114846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 05/06/2024] [Accepted: 06/06/2024] [Indexed: 07/30/2024]
Abstract
Pain in Parkinson's disease (PD) has been validated as one of the major non-motor dysfunctions affecting the quality of life and subsequent rehabilitation. In the present study, we investigated the role of the dopamine D3 receptor in the thalamic mediodorsal (MD) and ventromedial (VM) nuclei mediated descending control of nociception and intramuscular (i.m.) 2.5% formalin-induced persistent muscle nociception. Paw withdrawal reflexes were measured in naive rats and rats subjected to PD induced by unilateral microinjection of 6 μg 6-OHDA into the rat striatum. Formalin-induced muscle nociception in phase 1, inter-phase, and phase 2 was significantly greater in PD rats compared to naive and vehicle-treated rats (P < 0.001). PD rats exhibited bilaterally mechanical hyperalgesia and heat hypoalgesia in formalin-induced muscle nociception. Microinjection of SK609, a dopamine D3 receptor agonist, at various doses (2.5-7.5 nmol/0.5 μl) into the thalamic VM nucleus dose-dependently prolonged heat-evoked paw withdrawal latencies in both naive and PD rats. Administration of SK609 to either the MD or VM nuclei had no effect on noxious mechanically evoked paw withdrawal reflexes. Pre-treatment of the thalamic MD nucleus with SK609 significantly attenuated formalin-induced nociception, and reversed mechanical hyperalgesia, but not heat hypoalgesia. Pre-treatment of the thalamic VM nucleus with SK609 inhibited formalin-induced nociception in the late phase of phase 2 (30-75 min) and heat hypoalgesia, but not mechanical hyperalgesia (P < 0.05). It is suggested that the dopamine D3 receptors in the thalamus play an antinociceptive role in the descending modulation of nociception. Activation of D3 receptors within the thalamic MD and VM nuclei attenuates descending facilitation and enhances descending inhibition in rats during PD.
Collapse
Affiliation(s)
- Jing Lei
- Center for Translational Medicine Research on Sensory-Motor Diseases, Yan'an University, Yan'an 716000, PR China; Key Laboratory of Yan'an Sports Rehabilitation Medicine, Yan'an 716000, PR China
| | - Lin-Lin Tang
- Center for Translational Medicine Research on Sensory-Motor Diseases, Yan'an University, Yan'an 716000, PR China; Key Laboratory of Yan'an Sports Rehabilitation Medicine, Yan'an 716000, PR China
| | - Rong Jing
- Department of Rehabilitation Medicine, Affiliated Hospital of Yan'an University, Yan'an 716000, PR China
| | - Hao-Jun You
- Center for Translational Medicine Research on Sensory-Motor Diseases, Yan'an University, Yan'an 716000, PR China; Key Laboratory of Yan'an Sports Rehabilitation Medicine, Yan'an 716000, PR China.
| |
Collapse
|
4
|
Kubota H, Zhou X, Zhang X, Watanabe H, Nagai T. Pramipexole Hyperactivates the External Globus Pallidus and Impairs Decision-Making in a Mouse Model of Parkinson's Disease. Int J Mol Sci 2024; 25:8849. [PMID: 39201535 PMCID: PMC11354263 DOI: 10.3390/ijms25168849] [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: 07/18/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
In patients with Parkinson's disease (PD), dopamine replacement therapy with dopamine D2/D3 receptor agonists induces impairments in decision-making, including pathological gambling. The neurobiological mechanisms underlying these adverse effects remain elusive. Here, in a mouse model of PD, we investigated the effects of the dopamine D3 receptor (D3R)-preferring agonist pramipexole (PPX) on decision-making. PD model mice were generated using a bilateral injection of the toxin 6-hydroxydopamine into the dorsolateral striatum. Subsequent treatment with PPX increased disadvantageous choices characterized by a high-risk/high-reward in the touchscreen-based Iowa Gambling Task. This effect was blocked by treatment with the selective D3R antagonist PG-01037. In model mice treated with PPX, the number of c-Fos-positive cells was increased in the external globus pallidus (GPe), indicating dysregulation of the indirect pathway in the corticothalamic-basal ganglia circuitry. In accordance, chemogenetic inhibition of the GPe restored normal c-Fos activation and rescued PPX-induced disadvantageous choices. These findings demonstrate that the hyperactivation of GPe neurons in the indirect pathway impairs decision-making in PD model mice. The results provide a candidate mechanism and therapeutic target for pathological gambling observed during D2/D3 receptor pharmacotherapy in PD patients.
Collapse
Affiliation(s)
- Hisayoshi Kubota
- Division of Behavioral Neuropharmacology, International Center for Brain Science (ICBS), Fujita Health University, Toyoake 470-1192, Aichi, Japan; (H.K.)
| | - Xinzhu Zhou
- Division of Behavioral Neuropharmacology, International Center for Brain Science (ICBS), Fujita Health University, Toyoake 470-1192, Aichi, Japan; (H.K.)
| | - Xinjian Zhang
- Division of Behavioral Neuropharmacology, International Center for Brain Science (ICBS), Fujita Health University, Toyoake 470-1192, Aichi, Japan; (H.K.)
| | - Hirohisa Watanabe
- Department of Neurology, School of Medicine, Fujita Health University, Toyoake 470-1192, Aichi, Japan
| | - Taku Nagai
- Division of Behavioral Neuropharmacology, International Center for Brain Science (ICBS), Fujita Health University, Toyoake 470-1192, Aichi, Japan; (H.K.)
| |
Collapse
|
5
|
Zeng X, Wu C, Cao Y, Li H, Zhang X. Mdm2-mediated ubiquitination of PKCβII is responsible for insulin-induced heterologous desensitization of dopamine D 3 receptor. FEBS Lett 2024; 598:400-414. [PMID: 38302840 DOI: 10.1002/1873-3468.14815] [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: 11/13/2023] [Revised: 12/21/2023] [Accepted: 01/05/2024] [Indexed: 02/03/2024]
Abstract
The insulin and dopaminergic systems in the brain are associated with schizophrenia and Parkinson's disease with respect to etiology and treatment. The present study investigated the crosstalk between the insulin receptor (IR) and dopamine receptor and found that insulin stimulation selectively inhibits signaling of D3 R in a PKCβII-dependent manner. Upon insulin stimulation, E3 ligase enzyme Mdm2 moves out of the nucleus to ubiquitinate PKCβII. Subsequently, ubiquitinated PKCβII translocates to the cell membrane and interacts with D3 R in a phosphorylation-dependent manner at S229/257, resulting in the attenuation of D3 R signaling and initiating clathrin-mediated endocytosis and downregulation. Considering that both IR and D3 R are closely related to some neuropsychosis, this study could provide new molecular insight into the etiology of the disorder.
Collapse
Affiliation(s)
- Xingyue Zeng
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| | - ChengYan Wu
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| | - Yongkai Cao
- Department of Neurology, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, China
| | - Huijun Li
- Department of Pharmaceuticals, People's Hospital of Zunyi City Bo Zhou District, China
| | - Xiaohan Zhang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang, China
| |
Collapse
|
6
|
Alvarez-Herrera S, Rosel Vales M, Pérez-Sánchez G, Becerril-Villanueva E, Flores-Medina Y, Maldonado-García JL, Saracco-Alvarez R, Escamilla R, Pavón L. Risperidone Decreases Expression of Serotonin Receptor-2A (5-HT2A) and Serotonin Transporter (SERT) but Not Dopamine Receptors and Dopamine Transporter (DAT) in PBMCs from Patients with Schizophrenia. Pharmaceuticals (Basel) 2024; 17:167. [PMID: 38399382 PMCID: PMC10892557 DOI: 10.3390/ph17020167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 02/25/2024] Open
Abstract
Dopamine and serotonin receptors and transporters play an essential role in the pathophysiology of schizophrenia; changes in their expression have been reported in neurons and leukocytes. Each antipsychotic induces a unique pattern in leukocyte function and phenotype. However, the use of polytherapy to treat schizophrenia makes it challenging to determine the specific effects of risperidone on peripheral blood mononuclear cells (PBMCs). The aim of this study was to evaluate the changes in the expression of D3, D5, DAT, 5-HT2A, and SERT in PBMCs from healthy volunteers (HV), drug-naive patients with schizophrenia (PWS), drug-free PWS, and PWS treated with risperidone for up to 40 weeks using quantitative PCR. Our study revealed elevated mRNA levels of D3, DAT, 5-HT2A, and SERT in unmedicated PWS. Treatment with risperidone led to a reduction only in the expression of 5-HT2A and SERT. Furthermore, we observed a moderate correlation between 5-HT2A expression and the positive and negative syndrome scale (PANSS), as well as SERT expression and PANSS scale. We also found a moderate correlation between 5-HT2A and SERT expression and the positive subscale. The duration of risperidone consumption had a significant negative correlation with the expression of 5-HT2A and SERT. Our study introduces the measurement of 5-HT2A and SERT expression in PBMCs as a useful parameter for assessing the response to risperidone in PWS.
Collapse
Affiliation(s)
- Samantha Alvarez-Herrera
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñíz, Mexico City 14370, Mexico; (S.A.-H.); (G.P.-S.); (E.B.-V.)
| | - Mauricio Rosel Vales
- Clínica de Esquizofrenia, Dirección de Servicios Clínicos, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñíz, Mexico City 14370, Mexico;
| | - Gilberto Pérez-Sánchez
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñíz, Mexico City 14370, Mexico; (S.A.-H.); (G.P.-S.); (E.B.-V.)
| | - Enrique Becerril-Villanueva
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñíz, Mexico City 14370, Mexico; (S.A.-H.); (G.P.-S.); (E.B.-V.)
| | - Yvonne Flores-Medina
- Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñíz, Mexico City 14370, Mexico; (Y.F.-M.); (R.S.-A.)
| | - José Luis Maldonado-García
- Departamemto de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico;
- Departamemto de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Ricardo Saracco-Alvarez
- Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñíz, Mexico City 14370, Mexico; (Y.F.-M.); (R.S.-A.)
| | - Raúl Escamilla
- Subdirección de Consulta Externa, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñíz, Mexico City 14370, Mexico;
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Dirección de Investigaciones en Neurociencias, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñíz, Mexico City 14370, Mexico; (S.A.-H.); (G.P.-S.); (E.B.-V.)
| |
Collapse
|
7
|
Casados-Delgado O, Avalos-Fuentes JA, Lara-Lozano M, Tovar-Medina G, Florán-Hernández CD, Martínez-Nolasco KG, Cortes H, Felix R, Segovia J, Florán B. Modulation of D 3R Splicing, Signaling, and Expression by D 1R through PKA→PTB Phosphorylation. Biomedicines 2024; 12:206. [PMID: 38255311 PMCID: PMC10813448 DOI: 10.3390/biomedicines12010206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
The D1R and D3R receptors functionally and synergistically interact in striatonigral neurons. Dopaminergic denervation turns this interaction antagonistic, which is correlated with a decrement in D3nf isoform and an increment in D3R membranal expression. The mechanisms of such changes in D3R are attributed to the dysregulation of the expression of their isoforms. The cause and mechanism of this phenomenon remain unknown. Dopaminergic denervation produces a decrement in D1R and PKA activity; we propose that the lack of phosphorylation of PTB (regulator of alternative splicing) by PKA produces the dysregulation of D3R splicing and changes D3R functionality. By using in silico analysis, we found that D3R mRNA has motifs for PTB binding and, by RIP, co-precipitates with PTB. Moreover, D1R activation via PKA promotes PTB phosphorylation. Acute and 5-day D1R blockade decreases the expression of D3nf mRNA. The 5-day treatment reduces D3R, D3nf, and PTB protein in the cytoplasm and increases D3R in the membrane and PTB in the nucleus. Finally, the blockade of D1R mimics the effect of dopaminergic denervation in D1R and D3R signaling. Thus, our data indicate that through PKA→PTB, D1R modulates D3R splicing, expression, and signaling, which are altered during D1R blockade or the lack of stimulation in dopaminergic denervation.
Collapse
Affiliation(s)
- Orlando Casados-Delgado
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (O.C.-D.); (J.A.A.-F.); (M.L.-L.); (G.T.-M.); (C.D.F.-H.); (K.G.M.-N.); (J.S.)
| | - José Arturo Avalos-Fuentes
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (O.C.-D.); (J.A.A.-F.); (M.L.-L.); (G.T.-M.); (C.D.F.-H.); (K.G.M.-N.); (J.S.)
| | - Manuel Lara-Lozano
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (O.C.-D.); (J.A.A.-F.); (M.L.-L.); (G.T.-M.); (C.D.F.-H.); (K.G.M.-N.); (J.S.)
| | - Gisela Tovar-Medina
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (O.C.-D.); (J.A.A.-F.); (M.L.-L.); (G.T.-M.); (C.D.F.-H.); (K.G.M.-N.); (J.S.)
| | - Carla Daniela Florán-Hernández
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (O.C.-D.); (J.A.A.-F.); (M.L.-L.); (G.T.-M.); (C.D.F.-H.); (K.G.M.-N.); (J.S.)
| | - Karla Gisela Martínez-Nolasco
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (O.C.-D.); (J.A.A.-F.); (M.L.-L.); (G.T.-M.); (C.D.F.-H.); (K.G.M.-N.); (J.S.)
| | - Hernán Cortes
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Mexico City 14389, Mexico;
| | - Ricardo Felix
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico;
| | - José Segovia
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (O.C.-D.); (J.A.A.-F.); (M.L.-L.); (G.T.-M.); (C.D.F.-H.); (K.G.M.-N.); (J.S.)
| | - Benjamín Florán
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico; (O.C.-D.); (J.A.A.-F.); (M.L.-L.); (G.T.-M.); (C.D.F.-H.); (K.G.M.-N.); (J.S.)
| |
Collapse
|
8
|
Belloso-Iguerategui A, Zamarbide M, Merino-Galan L, Rodríguez-Chinchilla T, Gago B, Santamaria E, Fernández-Irigoyen J, Cotman CW, Prieto GA, Quiroga-Varela A, Rodríguez-Oroz MC. Hippocampal synaptic failure is an early event in experimental parkinsonism with subtle cognitive deficit. Brain 2023; 146:4949-4963. [PMID: 37403195 PMCID: PMC10690043 DOI: 10.1093/brain/awad227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/25/2023] [Accepted: 06/20/2023] [Indexed: 07/06/2023] Open
Abstract
Learning and memory mainly rely on correct synaptic function in the hippocampus and other brain regions. In Parkinson's disease, subtle cognitive deficits may even precede motor signs early in the disease. Hence, we set out to unravel the earliest hippocampal synaptic alterations associated with human α-synuclein overexpression prior to and soon after the appearance of cognitive deficits in a parkinsonism model. We bilaterally injected adeno-associated viral vectors encoding A53T-mutated human α-synuclein into the substantia nigra of rats, and evaluated them 1, 2, 4 and 16 weeks post-inoculation by immunohistochemistry and immunofluorescence to study degeneration and distribution of α-synuclein in the midbrain and hippocampus. The object location test was used to evaluate hippocampal-dependent memory. Sequential window acquisition of all theoretical mass spectrometry-based proteomics and fluorescence analysis of single-synapse long-term potentiation were used to study alterations to protein composition and plasticity in isolated hippocampal synapses. The effect of L-DOPA and pramipexole on long-term potentiation was also tested. Human α-synuclein was found within dopaminergic and glutamatergic neurons of the ventral tegmental area, and in dopaminergic, glutamatergic and GABAergic axon terminals in the hippocampus from 1 week post-inoculation, concomitant with mild dopaminergic degeneration in the ventral tegmental area. In the hippocampus, differential expression of proteins involved in synaptic vesicle cycling, neurotransmitter release and receptor trafficking, together with impaired long-term potentiation were the first events observed (1 week post-inoculation), preceding cognitive deficits (4 weeks post-inoculation). Later on, at 16 weeks post-inoculation, there was a deregulation of proteins involved in synaptic function, particularly those involved in the regulation of membrane potential, ion balance and receptor signalling. Hippocampal long-term potentiation was impaired before and soon after the onset of cognitive deficits, at 1 and 4 weeks post-inoculation, respectively. L-DOPA recovered hippocampal long-term potentiation more efficiently at 4 weeks post-inoculation than pramipexole, which partially rescued it at both time points. Overall, we found impaired synaptic plasticity and proteome dysregulation at hippocampal terminals to be the first events that contribute to the development of cognitive deficits in experimental parkinsonism. Our results not only point to dopaminergic but also to glutamatergic and GABAergic dysfunction, highlighting the relevance of the three neurotransmitter systems in the ventral tegmental area-hippocampus interaction from the earliest stages of parkinsonism. The proteins identified in the current work may constitute potential biomarkers of early synaptic damage in the hippocampus and hence, therapies targeting these could potentially restore early synaptic malfunction and consequently, cognitive deficits in Parkinson's disease.
Collapse
Affiliation(s)
| | - Marta Zamarbide
- Neuroscience Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, 31008 Pamplona, Spain
| | - Leyre Merino-Galan
- Neuroscience Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, 31008 Pamplona, Spain
- Neuroscience Department, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | | | - Belén Gago
- Faculty of Medicine, IBIMA Plataforma BIONAND, Universidad de Málaga, 29016 Málaga, Spain
| | - Enrique Santamaria
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain
- Neurosciences and Mental Health Area, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Unit, Proteomics Platform, Proteored-ISCIII, Navarrabiomed, Complejo Hospitalario de Navarra (CHN), Universidad Pública de Navarra (UPNA), 31008 Pamplona, Spain
- Neurosciences and Mental Health Area, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - Carl W Cotman
- Institute for Memory Impairments and Neurological Disorders, University of California-Irvine, Irvine, CA 92697, USA
| | - G Aleph Prieto
- Institute for Memory Impairments and Neurological Disorders, University of California-Irvine, Irvine, CA 92697, USA
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, 76010 Querétaro, México
| | - Ana Quiroga-Varela
- Neuroscience Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, 31008 Pamplona, Spain
- Neurosciences and Mental Health Area, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
| | - María Cruz Rodríguez-Oroz
- Neuroscience Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, 31008 Pamplona, Spain
- Neurosciences and Mental Health Area, Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain
- Neurology Department, Clínica Universidad de Navarra (CUN), 31008 Pamplona, Spain
| |
Collapse
|
9
|
Tall P, Qamar MA, Rosenzweig I, Raeder V, Sauerbier A, Heidemarie Z, Falup-Pecurariu C, Chaudhuri KR. The Park Sleep subtype in Parkinson's disease: from concept to clinic. Expert Opin Pharmacother 2023; 24:1725-1736. [PMID: 37561080 DOI: 10.1080/14656566.2023.2242786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 07/27/2023] [Indexed: 08/11/2023]
Abstract
INTRODUCTION The heterogeneity of Parkinson's disease (PD) is evident from descriptions of non-motor (NMS) subtypes and Park Sleep, originally identified by Sauerbier et al. 2016, is one such clinical subtype associated with the predominant clinical presentation of sleep dysfunctions including excessive daytime sleepiness (EDS), along with insomnia. AREAS COVERED A literature search was conducted using the PubMed, Medline, Embase, and Web of Science databases, accessed between 1 February 2023 and 28 March 2023. In this review, we describe the clinical subtype of Park Sleep and related 'tests' ranging from polysomnography to investigational neuromelanin MRI brain scans and some tissue-based biological markers. EXPERT OPINION Cholinergic, noradrenergic, and serotonergic systems are dominantly affected in PD. Park Sleep subtype is hypothesized to be associated primarily with serotonergic deficit, clinically manifesting as somnolence and narcoleptic events (sleep attacks), with or without rapid eye movement behavior disorder (RBD). In clinic, Park Sleep recognition may drive lifestyle changes (e.g. driving) along with therapy adjustments as Park Sleep patients may be sensitive to dopamine D3 active agonists, such as ropinirole and pramipexole. Specific dashboard scores based personalized management options need to be implemented and include pharmacological, non-pharmacological, and lifestyle linked advice.
Collapse
Affiliation(s)
- Phoebe Tall
- Department of Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience (IoPpn), King's College London, London, UK
- Parkinson's Foundation Centre of Excellence, King's College Hospital NHS Foundation Trust, London, UK
| | - Mubasher A Qamar
- Department of Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience (IoPpn), King's College London, London, UK
- Parkinson's Foundation Centre of Excellence, King's College Hospital NHS Foundation Trust, London, UK
| | - Ivana Rosenzweig
- Sleep and Brain Plasticity Centre, Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPpn), King's College London, London, UK
- Sleep Disorder Centre, Nuffield House, Guy's Hospital, London, UK
| | - Vanessa Raeder
- Parkinson's Foundation Centre of Excellence, King's College Hospital NHS Foundation Trust, London, UK
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität, Berlin, Germany
| | - Anna Sauerbier
- Department of Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience (IoPpn), King's College London, London, UK
- Department of Neurology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Zach Heidemarie
- Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Cristian Falup-Pecurariu
- Faculty of Medicine, Transilvania University of Braşov, Brașov, Romania
- Department of Neurology, County Clinic Hospital, Braşov, Romania
| | - Kallol Ray Chaudhuri
- Department of Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience (IoPpn), King's College London, London, UK
- Parkinson's Foundation Centre of Excellence, King's College Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
10
|
Gonçalves FB, Garcia-Gomes MSA, Silva-Sampaio AC, Kirsten TB, Bondan EF, Sandini TM, Flório JC, Lebrun I, Coque ADC, Alexandre-Ribeiro SR, Massironi SMG, Mori CMC, Bernardi MM. Progressive tremor and motor impairment in seizure-prone mutant tremor mice are associated with neurotransmitter dysfunction. Behav Brain Res 2023; 443:114329. [PMID: 36746310 DOI: 10.1016/j.bbr.2023.114329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND The tremor mutant mice present motor impairments comprised of whole-body tremors, ataxia, decreased exploratory behavior, and audiogenic seizures. OBJECTIVES This study aims to investigate the development of motor dysfunction in this mutant mouse and the relationships with cortical, striatal, and cerebellar levels of GABA, glutamate, glycine, dopamine (DA), serotonin (5-HT), noradrenaline (NOR), and its metabolites. The serum cytokines levels, myelin content, and the astrocytic expression of the glial fibrillary acidic protein (GFAP) investigated the possible influence of inflammation in motor dysfunction. RESULTS Relative to wild-type (WT) mice, the tremor mice presented: increased tremors and bradykinesia associated with postural instability, decreased range of motion, and difficulty in initiating voluntary movements directly proportional to age; reduced step length for right and left hindlimbs; reduced cortical GABA, glutamate and, aspartate levels, the DOPAC/DA and ratio and increased the NOR levels; in the striatum, the levels of glycine and aspartate were reduced while the HVA levels, the HVA/DA and 5HIAA/5-HT ratios increased; in the cerebellum the glycine, NOR and 5-HIAA levels increased. CONCLUSIONS We suggest that the motor disturbances resulted mainly from the activation of the indirect striatal inhibitory pathway to the frontal cortex mediated by GABA, glutamate, and aspartate, reducing the dopaminergic activity at the prefrontal cortex, which was associated with the progressive tremor. The reduced striatal and increased cerebellar glycine levels could be partially responsible for the mutant tremor motor disturbances.
Collapse
Affiliation(s)
- Flávio B Gonçalves
- Psychoneuroimmunology Laboratory, Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Mariana S A Garcia-Gomes
- Department of Psychiatric, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Ana Claudia Silva-Sampaio
- Psychoneuroimmunology Laboratory, Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Thiago B Kirsten
- Psychoneuroimmunology Laboratory, Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Eduardo F Bondan
- Psychoneuroimmunology Laboratory, Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | - Thaísa M Sandini
- Department of Psychiatric, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Jorge C Flório
- Program in Experimental and Comparative Pathology, Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, São Paulo, SP 05508-270, Brazil
| | - Ivo Lebrun
- Laboratory of Biochemistry and Biophysics, Program in Toxinology, Butantan Institute, Brazil
| | - Alex de C Coque
- Psychoneuroimmunology Laboratory, Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil
| | | | - Silvia M G Massironi
- Program in Experimental and Comparative Pathology, Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, São Paulo, SP 05508-270, Brazil
| | - Claudia M C Mori
- Program in Experimental and Comparative Pathology, Department of Pathology, School of Veterinary Medicine and Animal Science, University of São Paulo, Av. Prof. Dr. Orlando Marques de Paiva, 87, São Paulo, SP 05508-270, Brazil
| | - Maria M Bernardi
- Psychoneuroimmunology Laboratory, Program in Environmental and Experimental Pathology, Paulista University, Rua Dr. Bacelar, 1212, São Paulo, SP 04026-002, Brazil.
| |
Collapse
|
11
|
Gogarnoiu ES, Vogt CD, Sanchez J, Bonifazi A, Saab E, Shaik AB, Soler-Cedeño O, Bi GH, Klein B, Xi ZX, Lane JR, Newman AH. Dopamine D 3/D 2 Receptor Ligands Based on Cariprazine for the Treatment of Psychostimulant Use Disorders That May Be Dual Diagnosed with Affective Disorders. J Med Chem 2023; 66:1809-1834. [PMID: 36661568 PMCID: PMC11100975 DOI: 10.1021/acs.jmedchem.2c01624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Highly selective dopamine D3 receptor (D3R) partial agonists/antagonists have been developed for the treatment of psychostimulant use disorders (PSUD). However, none have reached the clinic due to insufficient potency/efficacy or potential cardiotoxicity. Cariprazine, an FDA-approved drug for the treatment of schizophrenia and bipolar disorder, is a high-affinity D3R partial agonist (Ki = 0.22 nM) with 3.6-fold selectivity over the homologous dopamine D2 receptor (D2R). We hypothesized that compounds that are moderately D3R/D2R-selective partial agonists/antagonists may be effective for the treatment of PSUD. By systematically modifying the parent molecule, we discovered partial agonists/antagonists, as measured in bioluminescence resonance energy transfer (BRET)-based assays, with high D3R affinities (Ki = 0.14-50 nM) and moderate selectivity (<100-fold) over D2R. Cariprazine and two lead analogues, 13a and 13e, decreased cocaine self-administration (FR2; 1-10 mg/kg, i.p.) in rats, suggesting that partial agonists/antagonists with modest D3R/D2R selectivity may be effective in treating PSUD and potentially comorbidities with other affective disorders.
Collapse
Affiliation(s)
- Emma S. Gogarnoiu
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Caleb D. Vogt
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Julie Sanchez
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom
- Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, Midlands NG2 7AG, United Kingdom
| | - Alessandro Bonifazi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Elizabeth Saab
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Anver Basha Shaik
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Omar Soler-Cedeño
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Guo-Hua Bi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Benjamin Klein
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - Zheng-Xiong Xi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| | - J. Robert Lane
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom
- Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, Midlands NG2 7AG, United Kingdom
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse – Intramural Research Program, National Institutes of Health, 333 Cassell Drive, Baltimore, Maryland 21224, United States
| |
Collapse
|
12
|
Juza R, Musilek K, Mezeiova E, Soukup O, Korabecny J. Recent advances in dopamine D 2 receptor ligands in the treatment of neuropsychiatric disorders. Med Res Rev 2023; 43:55-211. [PMID: 36111795 DOI: 10.1002/med.21923] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 07/29/2022] [Accepted: 08/09/2022] [Indexed: 02/04/2023]
Abstract
Dopamine is a biologically active amine synthesized in the central and peripheral nervous system. This biogenic monoamine acts by activating five types of dopamine receptors (D1-5 Rs), which belong to the G protein-coupled receptor family. Antagonists and partial agonists of D2 Rs are used to treat schizophrenia, Parkinson's disease, depression, and anxiety. The typical pharmacophore with high D2 R affinity comprises four main areas, namely aromatic moiety, cyclic amine, central linker and aromatic/heteroaromatic lipophilic fragment. From the literature reviewed herein, we can conclude that 4-(2,3-dichlorophenyl), 4-(2-methoxyphenyl)-, 4-(benzo[b]thiophen-4-yl)-1-substituted piperazine, and 4-(6-fluorobenzo[d]isoxazol-3-yl)piperidine moieties are critical for high D2 R affinity. Four to six atoms chains are optimal for D2 R affinity with 4-butoxyl as the most pronounced one. The bicyclic aromatic/heteroaromatic systems are most frequently occurring as lipophilic appendages to retain high D2 R affinity. In this review, we provide a thorough overview of the therapeutic potential of D2 R modulators in the treatment of the aforementioned disorders. In addition, this review summarizes current knowledge about these diseases, with a focus on the dopaminergic pathway underlying these pathologies. Major attention is paid to the structure, function, and pharmacology of novel D2 R ligands, which have been developed in the last decade (2010-2021), and belong to the 1,4-disubstituted aromatic cyclic amine group. Due to the abundance of data, allosteric D2 R ligands and D2 R modulators from patents are not discussed in this review.
Collapse
Affiliation(s)
- Radomir Juza
- Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic
| | - Kamil Musilek
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Hradec Kralove, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Eva Mezeiova
- Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Ondrej Soukup
- Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Jan Korabecny
- Experimental Neurobiology, National Institute of Mental Health, Klecany, Czech Republic.,Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| |
Collapse
|
13
|
Xu J. Dopamine D3 Receptor in Parkinson Disease: A Prognosis Biomarker and an Intervention Target. Curr Top Behav Neurosci 2023; 60:89-107. [PMID: 35711029 PMCID: PMC10034716 DOI: 10.1007/7854_2022_373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Parkinson disease (PD) dementia, pathologically featured as nigrostriatal dopamine (DA) neuronal loss with motor and non-motor manifestations, leads to substantial disability and economic burden. DA therapy targets the DA D3 receptor (D3R) with high affinity and selectivity. The pathological involvement of D3R is evidenced as an effective biomarker for disease progression and DA agnostic interventions, with compensations of increased DA, decreased aggregates of α-synuclein (α-Syn), enhanced secretion of brain-derived neurotrophic factors (BDNF), attenuation of neuroinflammation and oxidative damage, and promoting neurogenesis in the brain. D3R also interacts with D1R to reduce PD-associated motor symptoms and alleviate the side effects of levodopa (L-DOPA) treatment. We recently found that DA D2 receptor (D2R) density decreases in the late-stage PDs, while high D3R or DA D1 receptor (D1R) + D3R densities in the postmortem PD brains correlate with survival advantages. These new essential findings warrant renewed investigations into the understanding of D3R neuron populations and their cross-sectional and longitudinal regulations in PD progression.
Collapse
Affiliation(s)
- Jinbin Xu
- Division of Radiological Sciences, Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
14
|
Wei JR, Hao ZZ, Xu C, Huang M, Tang L, Xu N, Liu R, Shen Y, Teichmann SA, Miao Z, Liu S. Identification of visual cortex cell types and species differences using single-cell RNA sequencing. Nat Commun 2022; 13:6902. [PMID: 36371428 PMCID: PMC9653448 DOI: 10.1038/s41467-022-34590-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 10/31/2022] [Indexed: 11/13/2022] Open
Abstract
The primate neocortex exerts high cognitive ability and strong information processing capacity. Here, we establish a single-cell RNA sequencing dataset of 133,454 macaque visual cortical cells. It covers major cortical cell classes including 25 excitatory neuron types, 37 inhibitory neuron types and all glial cell types. We identified layer-specific markers including HPCAL1 and NXPH4, and also identified two cell types, an NPY-expressing excitatory neuron type that expresses the dopamine receptor D3 gene; and a primate specific activity-dependent OSTN + sensory neuron type. Comparisons of our dataset with humans and mice show that the gene expression profiles differ between species in relation to genes that are implicated in the synaptic plasticity and neuromodulation of excitatory neurons. The comparisons also revealed that glutamatergic neurons may be more diverse across species than GABAergic neurons and non-neuronal cells. These findings pave the way for understanding how the primary cortex fulfills the high-cognitive functions.
Collapse
Affiliation(s)
- Jia-Ru Wei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Zhao-Zhe Hao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Chuan Xu
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Mengyao Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Lei Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Nana Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Ruifeng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yuhui Shen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK.
- Department of Physics, Cavendish Laboratory, University of Cambridge, Cambridge, UK.
| | - Zhichao Miao
- GMU-GIBH Joint School of Life Sciences, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China.
- European Bioinformatics Institute, European Molecular Biology Laboratory, Wellcome Genome Campus, Cambridge, UK.
| | - Sheng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou, China.
| |
Collapse
|
15
|
Kato M, Kunisawa N, Shimizu S, Iha HA, Ohno Y. Mechanisms Underlying Dopaminergic Regulation of Nicotine-Induced Kinetic Tremor. Front Pharmacol 2022; 13:938175. [PMID: 35784764 PMCID: PMC9243423 DOI: 10.3389/fphar.2022.938175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Nicotine induces kinetic tremor, which resembles pharmacological features of essential tremors, via activating the inferior olive (IO) neurons. Since nicotine is known to enhance dopamine release by stimulating α4β2 and/or α6 nACh receptors, we examined the effects of various dopamine receptor ligands on nicotine-induced tremor to clarify the role of the dopaminergic system in modulating nicotine tremor. A tremorgenic dose of nicotine increased the dopamine level in the pons and medulla oblongata (P/MO), and the levels of dopamine metabolites in the hippocampus, P/MO, and striatum. Treatment of animals with the D1/5 agonist SKF-38393 inhibited the induction of nicotine tremor, whereas the D3 agonist PD-128,907 facilitated nicotine-induced tremor. The D2 agonist sumanirole showed no effect. In addition, nicotine tremor was significantly enhanced by the D1/5 antagonist SCH-23390 and inhibited by the D3 antagonist U-99194. Neither the D2 (L-741,626) nor D4 (L-745,870) antagonist affected the generation of nicotine tremor. Furthermore, microinjection of U-99194 into the cerebellum significantly inhibited nicotine-induced tremor, whereas its injection into IO or the striatum did not affect tremor generation. Although intrastriatal injection of SCH-23390 showed no effects, its injection into IO tended to enhance nicotine-induced tremor. The present study suggests that dopamine D3 and D1/5 receptors regulate the induction of nicotine tremor in an opposite way, D3 receptors facilitately and D1/5 receptors inhibitorily. In addition, the cerebellar D3 receptors may play an important role in modulating the induction of nicotine tremor mediated by the olivo-cerebellar system.
Collapse
|
16
|
Gondré-Lewis MC, Elman I, Alim T, Chapman E, Settles-Reaves B, Galvao C, Gold MS, Baron D, Kazmi S, Gardner E, Gupta A, Dennen C, Blum K. Frequency of the Dopamine Receptor D3 (rs6280) vs. Opioid Receptor µ1 (rs1799971) Polymorphic Risk Alleles in Patients with Opioid Use Disorder: A Preponderance of Dopaminergic Mechanisms? Biomedicines 2022; 10:870. [PMID: 35453620 PMCID: PMC9027142 DOI: 10.3390/biomedicines10040870] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/21/2022] [Accepted: 03/26/2022] [Indexed: 02/05/2023] Open
Abstract
While opioids are a powerful class of drugs that inhibit transmission of pain signals, their use is tarnished by the current epidemic of opioid use disorder (OUD) and overdose deaths. Notwithstanding published reports, there remain gaps in our knowledge of opioid receptor mechanisms and their role in opioid seeking behavior. Thus, novel insights into molecular, neurogenetic and neuropharmacological bases of OUD are needed. We propose that an addictive endophenotype may not be entirely specific to the drug of choice but rather may be generalizable to altered brain reward circuits impacting net mesocorticolimbic dopamine release. We suggest that genetic or epigenetic alterations across dopaminergic reward systems lead to uncontrollable self-administration of opioids and other drugs. For instance, diminished availability via knockout of dopamine D3 receptor (DRD3) increases vulnerability to opioids. Building upon this concept via the use of a sophisticated polymorphic risk analysis in a human cohort of chronic opioid users, we found evidence for a higher frequency of polymorphic DRD3 risk allele (rs6280) than opioid receptor µ1 (rs1799971). In conclusion, while opioidergic mechanisms are involved in OUD, dopamine-related receptors may have primary influence on opioid-seeking behavior in African Americans. These findings suggest OUD-targeted novel and improved neuropharmacological therapies may require focus on DRD3-mediated regulation of dopaminergic homeostasis.
Collapse
Affiliation(s)
- Marjorie C. Gondré-Lewis
- Neuropsychopharmacology Laboratory, Department of Anatomy, Howard University College of Medicine, Washington, DC 20059, USA (C.G.)
| | - Igor Elman
- Department of Psychiatry, Cambridge Health Alliance/Harvard Medical School, Cambridge, MA 02139, USA or
| | - Tanya Alim
- Department of Psychiatry and Behavioral Sciences, Howard University College of Medicine, Washington, DC 20059, USA; (T.A.); (E.C.)
| | - Edwin Chapman
- Department of Psychiatry and Behavioral Sciences, Howard University College of Medicine, Washington, DC 20059, USA; (T.A.); (E.C.)
| | - Beverlyn Settles-Reaves
- Neuropsychopharmacology Laboratory, Department of Anatomy, Howard University College of Medicine, Washington, DC 20059, USA (C.G.)
| | - Carine Galvao
- Neuropsychopharmacology Laboratory, Department of Anatomy, Howard University College of Medicine, Washington, DC 20059, USA (C.G.)
| | - Mark S. Gold
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - David Baron
- Graduate College, Western University Health Sciences, Pomona, CA 91766, USA;
| | - Shan Kazmi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA or
| | - Eliot Gardner
- Neuropsychopharmacology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA;
| | - Ashim Gupta
- Future Biologics, Lawrenceville, GA 30043, USA;
| | - Catherine Dennen
- The Kenneth Blum Behavioral & Neurogenetic Institute, Austin, TX 78701, USA;
| | - Kenneth Blum
- Graduate College, Western University Health Sciences, Pomona, CA 91766, USA;
- The Kenneth Blum Behavioral & Neurogenetic Institute, Austin, TX 78701, USA;
- Department of Clinical Psychology and Addiction, Institute of Psychology, Faculty of Education and Psychology, ELTE Eötvös Loránd University, Egyetem tér 1-3, 1053 Budapest, Hungary
- Department of Psychiatry, School of Medicine, University of Vermont, Burlington, VT 05405, USA
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology, Nonakuri, Purba Medinipur 721172, West Bengal, India
- Department of Psychiatry, Wright State University Boonshoft School of Medicine and Dayton VA Medical Centre, Dayton, OH 45324, USA
| |
Collapse
|
17
|
Jordan CJ, Xi ZX. Identification of the Risk Genes Associated With Vulnerability to Addiction: Major Findings From Transgenic Animals. Front Neurosci 2022; 15:811192. [PMID: 35095405 PMCID: PMC8789752 DOI: 10.3389/fnins.2021.811192] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/14/2021] [Indexed: 12/21/2022] Open
Abstract
Understanding risk factors for substance use disorders (SUD) can facilitate medication development for SUD treatment. While a rich literature exists discussing environmental factors that influence SUD, fewer articles have focused on genetic factors that convey vulnerability to drug use. Methods to identify SUD risk genes include Genome-Wide Association Studies (GWAS) and transgenic approaches. GWAS have identified hundreds of gene variants or single nucleotide polymorphisms (SNPs). However, few genes identified by GWAS have been verified by clinical or preclinical studies. In contrast, significant progress has been made in transgenic approaches to identify risk genes for SUD. In this article, we review recent progress in identifying candidate genes contributing to drug use and addiction using transgenic approaches. A central hypothesis is if a particular gene variant (e.g., resulting in reduction or deletion of a protein) is associated with increases in drug self-administration or relapse to drug seeking, this gene variant may be considered a risk factor for drug use and addiction. Accordingly, we identified several candidate genes such as those that encode dopamine D2 and D3 receptors, mGluR2, M4 muscarinic acetylcholine receptors, and α5 nicotinic acetylcholine receptors, which appear to meet the risk-gene criteria when their expression is decreased. Here, we describe the role of these receptors in drug reward and addiction, and then summarize major findings from the gene-knockout mice or rats in animal models of addiction. Lastly, we briefly discuss future research directions in identifying addiction-related risk genes and in risk gene-based medication development for the treatment of addiction.
Collapse
Affiliation(s)
- Chloe J. Jordan
- Division of Alcohol, Drugs and Addiction, Department of Psychiatry, McLean Hospital, Harvard Medical School, Belmont, MA, United States
- *Correspondence: Chloe J. Jordan,
| | - Zheng-Xiong Xi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, United States
- Zheng-Xiong Xi,
| |
Collapse
|
18
|
Kiss B, Krámos B, Laszlovszky I. Potential Mechanisms for Why Not All Antipsychotics Are Able to Occupy Dopamine D 3 Receptors in the Brain in vivo. Front Psychiatry 2022; 13:785592. [PMID: 35401257 PMCID: PMC8987915 DOI: 10.3389/fpsyt.2022.785592] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 02/25/2022] [Indexed: 11/29/2022] Open
Abstract
Dysfunctions of the dopaminergic system are believed to play a major role in the core symptoms of schizophrenia such as positive, negative, and cognitive symptoms. The first line of treatment of schizophrenia are antipsychotics, a class of medications that targets several neurotransmitter receptors in the brain, including dopaminergic, serotonergic, adrenergic and/or muscarinic receptors, depending on the given agent. Although the currently used antipsychotics display in vitro activity at several receptors, majority of them share the common property of having high/moderate in vitro affinity for dopamine D2 receptors (D2Rs) and D3 receptors (D3Rs). In terms of mode of action, these antipsychotics are either antagonist or partial agonist at the above-mentioned receptors. Although D2Rs and D3Rs possess high degree of homology in their molecular structure, have common signaling pathways and similar in vitro pharmacology, they have different in vivo pharmacology and therefore behavioral roles. The aim of this review, with summarizing preclinical and clinical evidence is to demonstrate that while currently used antipsychotics display substantial in vitro affinity for both D3Rs and D2Rs, only very few can significantly occupy D3Rs in vivo. The relative importance of the level of endogenous extracellular dopamine in the brain and the degree of in vitro D3Rs receptor affinity and selectivity as determinant factors for in vivo D3Rs occupancy by antipsychotics, are also discussed.
Collapse
Affiliation(s)
- Béla Kiss
- Pharmacological and Drug Safety Research, Gedeon Richter Plc., Budapest, Hungary
| | - Balázs Krámos
- Spectroscopic Research Department, Gedeon Richter Plc., Budapest, Hungary
| | | |
Collapse
|
19
|
Lanza K, Bishop C. Dopamine D3 Receptor Plasticity in Parkinson's Disease and L-DOPA-Induced Dyskinesia. Biomedicines 2021; 9:biomedicines9030314. [PMID: 33808538 PMCID: PMC8003204 DOI: 10.3390/biomedicines9030314] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 11/21/2022] Open
Abstract
Parkinson’s Disease (PD) is characterized by primary and secondary plasticity that occurs in response to progressive degeneration and long-term L-DOPA treatment. Some of this plasticity contributes to the detrimental side effects associated with chronic L-DOPA treatment, namely L-DOPA-induced dyskinesia (LID). The dopamine D3 receptor (D3R) has emerged as a promising target in LID management as it is upregulated in LID. This upregulation occurs primarily in the D1-receptor-bearing (D1R) cells of the striatum, which have been repeatedly implicated in LID manifestation. D3R undergoes dynamic changes both in PD and in LID, making it difficult to delineate D3R’s specific contributions, but recent genetic and pharmacologic tools have helped to clarify its role in LID. The following review will discuss these changes, recent advances to better clarify D3R in both PD and LID and potential steps for translating these findings.
Collapse
Affiliation(s)
- Kathryn Lanza
- Department of Physiology, Northwestern University, Chicago, IL 60201, USA;
| | - Christopher Bishop
- Department of Psychology, Binghamton University, Binghamton, NY 13902, USA
- Correspondence:
| |
Collapse
|
20
|
Elek M, Djokovic N, Frank A, Oljacic S, Zivkovic A, Nikolic K, Stark H. Synthesis, in silico, and in vitro studies of novel dopamine D 2 and D 3 receptor ligands. Arch Pharm (Weinheim) 2021; 354:e2000486. [PMID: 33615541 DOI: 10.1002/ardp.202000486] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/30/2022]
Abstract
Dopamine is an important neurotransmitter in the human brain and its altered concentrations can lead to various neurological diseases. We studied the binding of novel compounds at the dopamine D2 (D2 R) and D3 (D3 R) receptor subtypes, which belong to the D2 -like receptor family. The synthesis, in silico, and in vitro characterization of 10 dopamine receptor ligands were performed. Novel ligands were docked into the D2 R and D3 R crystal structures to examine the precise binding mode. A quantum mechanics/molecular mechanics study was performed to gain insights into the nature of the intermolecular interactions between the newly introduced pentafluorosulfanyl (SF5 ) moiety and D2 R and D3 R. A radioligand displacement assay determined that all of the ligands showed moderate-to-low nanomolar affinities at D2 R and D3 R, with a slight preference for D3 R, which was confirmed in the in silico studies. N-{4-[4-(2-Methoxyphenyl)piperazin-1-yl]butyl}-4-(pentafluoro-λ6-sulfanyl)benzamide (7i) showed the highest D3 R affinity and selectivity (pKi values of 7.14 [D2 R] and 8.42 [D3 R]).
Collapse
Affiliation(s)
- Milica Elek
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitaetsstr. 1, Duesseldorf, NRW, Germany
| | - Nemanja Djokovic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Annika Frank
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitaetsstr. 1, Duesseldorf, NRW, Germany
| | - Slavica Oljacic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Zivkovic
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitaetsstr. 1, Duesseldorf, NRW, Germany
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitaetsstr. 1, Duesseldorf, NRW, Germany
| |
Collapse
|
21
|
Cosi C, Martel JC, Auclair AL, Collo G, Cavalleri L, Heusler P, Leriche L, Gaudoux F, Sokoloff P, Moser PC, Gatti-McArthur S. Pharmacology profile of F17464, a dopamine D 3 receptor preferential antagonist. Eur J Pharmacol 2021; 890:173635. [PMID: 33065094 DOI: 10.1016/j.ejphar.2020.173635] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 10/01/2020] [Accepted: 10/05/2020] [Indexed: 12/19/2022]
Abstract
F17464 (N-(3-{4-[4-(8-Oxo-8H-[1,3]-dioxolo-[4,5-g]-chromen-7-yl)-butyl]-piperazin-1-yl}-phenyl)-methanesulfonamide, hydrochloride) is a new potential antipsychotic with a unique profile. The compound exhibits high affinity for the human dopamine receptor subtype 3 (hD3) (Ki = 0.17 nM) and the serotonin receptor subtype 1a (5-HT1a) (Ki = 0.16 nM) and a >50 fold lower affinity for the human dopamine receptor subtype 2 short and long form (hD2s/l) (Ki = 8.9 and 12.1 nM, respectively). [14C]F17464 dynamic studies show a slower dissociation rate from hD3 receptor (t1/2 = 110 min) than from hD2s receptor (t1/2 = 1.4 min) and functional studies demonstrate that F17464 is a D3 receptor antagonist, 5-HT1a receptor partial agonist. In human dopaminergic neurons F17464 blocks ketamine induced morphological changes, an effect D3 receptor mediated. In vivo F17464 target engagement of both D2 and 5-HT1a receptors is demonstrated in displacement studies in the mouse brain. F17464 increases dopamine release in the rat prefrontal cortex and mouse lateral forebrain - dorsal striatum and seems to reduce the effect of MK801 on % c-fos mRNA medium expressing neurons in cortical and subcortical regions. F17464 also rescues valproate induced impairment in a rat social interaction model of autism. All the neurochemistry and behavioural effects of F17464 are observed in the dose range 0.32-2.5 mg/kg i.p. in both rats and mice. The in vitro - in vivo pharmacology profile of F17464 in preclinical models is discussed in support of a therapeutic use of the compound in schizophrenia and autism.
Collapse
Affiliation(s)
- Cristina Cosi
- Innovation Unit CNS, CEPC Pierre Fabre Laboratories, Bel Air de Campans, 81106, Castres, France
| | - Jean-Claude Martel
- Innovation Unit CNS, CEPC Pierre Fabre Laboratories, Bel Air de Campans, 81106, Castres, France
| | - Agnès L Auclair
- Innovation Unit CNS, CEPC Pierre Fabre Laboratories, Bel Air de Campans, 81106, Castres, France
| | - Ginetta Collo
- Dept of Molecular and Translational Medicine University of Brescia, Viale Europa 11, Brescia, Italy
| | - Laura Cavalleri
- Dept of Molecular and Translational Medicine University of Brescia, Viale Europa 11, Brescia, Italy
| | - Peter Heusler
- Innovation Unit CNS, CEPC Pierre Fabre Laboratories, Bel Air de Campans, 81106, Castres, France
| | - Ludovic Leriche
- Innovation Unit CNS, CEPC Pierre Fabre Laboratories, Bel Air de Campans, 81106, Castres, France
| | - Florence Gaudoux
- Innovation Unit CNS, CEPC Pierre Fabre Laboratories, Bel Air de Campans, 81106, Castres, France
| | - Pierre Sokoloff
- Innovation Unit CNS, CEPC Pierre Fabre Laboratories, Bel Air de Campans, 81106, Castres, France
| | - Paul C Moser
- Innovation Unit CNS, CEPC Pierre Fabre Laboratories, Bel Air de Campans, 81106, Castres, France
| | - Silvia Gatti-McArthur
- Innovation Unit CNS, CEPC Pierre Fabre Laboratories, Bel Air de Campans, 81106, Castres, France.
| |
Collapse
|
22
|
Paudel P, Shrestha S, Park SE, Seong SH, Fauzi FM, Jung HA, Choi JS. Emodin Derivatives as Multi-Target-Directed Ligands Inhibiting Monoamine Oxidase and Antagonizing Vasopressin V 1A Receptors. ACS OMEGA 2020; 5:26720-26731. [PMID: 33110998 PMCID: PMC7581273 DOI: 10.1021/acsomega.0c03649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/23/2020] [Indexed: 05/13/2023]
Abstract
The brain neurotransmitter level is associated with the pathology of various neurodegenerative diseases, and age-dependent increase in the blood level of vasopressin, human brain monoamine oxidase (hMAO) level, oxidative stress, and imbalance in aminergic signaling are common disease-modifying factors leading to various neurodegenerative disorders. Based on the reports of emodin in hMAO inhibition and antagonist effect on the vasopressin V1A receptor, in this study we synthesized six emodin derivatives and evaluated their effects on MAO activity and G protein-coupled receptors. Among them, 4-hydroxyemodin and 5-hydroxyemodin were potent inhibitors of hMAO, and 2-hydroxyemodin and 5-hydroxyemodin were good V1AR antagonists. In silico molecular docking simulation revealed that the hydroxyl group at C2, C4, and C5 of the respective compounds interacted with prime residues, which corroborates the in vitro effect. Likewise, these three derivatives were predicted to have good drug-like properties. Overall, our study demonstrates that the hydroxyl derivatives of emodin are multi-target-directed ligands that may act as leads for the design and development of a therapy for central nervous system disorders.
Collapse
Affiliation(s)
- Pradeep Paudel
- Department
of Food and Life Science, Pukyong National
University, Busan 48513, Republic of Korea
- National
Center for Natural Products Research, The
University of Mississippi, Oxford, Mississippi 38677, United States
| | - Srijan Shrestha
- Discipline
of Pharmacology, School of Medicine, Faculty of Health Science, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Se Eun Park
- Department
of Food and Life Science, Pukyong National
University, Busan 48513, Republic of Korea
| | - Su Hui Seong
- Department
of Food and Life Science, Pukyong National
University, Busan 48513, Republic of Korea
| | - Fazlin Mohd Fauzi
- Department
of Pharmacology and Chemistry, Faculty of Pharmacy, Universiti Teknologi MARA, Selangor Branch, Puncak Alam Campus, Bandar Puncak Alam, Selangor 42300, Malaysia
| | - Hyun Ah Jung
- Department
of Food Science and Human Nutrition, Jeonbuk
National University, Jeonju 54896, Republic of Korea
- . Phone: 82-63-270-4882. Fax: 82-63-270-3854
| | - Jae Sue Choi
- Department
of Food and Life Science, Pukyong National
University, Busan 48513, Republic of Korea
- . Phone: +82-51-629-5845. Fax: +82 51 629 5842
| |
Collapse
|
23
|
Saganuwan SA. Chemistry and Effects of Brainstem Acting Drugs. Cent Nerv Syst Agents Med Chem 2020; 19:180-186. [PMID: 31223094 DOI: 10.2174/1871524919666190620164355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 05/02/2019] [Accepted: 05/07/2019] [Indexed: 01/29/2023]
Abstract
BACKGROUND Brain is the most sensitive organ, whereas brainstem is the most important part of Central Nervous System (CNS). It connects the brain and the spinal cord. However, a myriad of drugs and chemicals affects CNS with severe resultant effects on the brainstem. METHODS In view of this, a number of literature were assessed for information on the most sensitive part of brain, drugs and chemicals that act on the brainstem and clinical benefit and risk assessment of such drugs and chemicals. RESULTS Findings have shown that brainstem regulates heartbeat, respiration and because it connects the brain and spinal cord, all the drugs that act on the spinal cord may overall affect the systems controlled by the spinal cord and brain. The message is sent and received by temporal lobe, occipital lobe, frontal lobe, parietal lobe and cerebellum. CONCLUSION Hence, the chemical functional groups of the brainstem and drugs acting on brainstem are complementary, and may produce either stimulation or depression of CNS.
Collapse
Affiliation(s)
- Saganuwan Alhaji Saganuwan
- Department of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Federal University of Agriculture, P.M.B. 2373, Makurdi, Benue State, Nigeria
| |
Collapse
|
24
|
Paudel P, Park SE, Seong SH, Jung HA, Choi JS. Bromophenols from Symphyocladia latiuscula Target Human Monoamine Oxidase and Dopaminergic Receptors for the Management of Neurodegenerative Diseases. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:2426-2436. [PMID: 32011134 DOI: 10.1021/acs.jafc.0c00007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Progressive degeneration of dopaminergic neurons in the substantia nigra is the characteristic feature of Parkinson's disease (PD) and the severity accelerates with aging. Therefore, improving dopamine level or dopamine receptor signaling is a standard approach for PD treatment. Herein, our results demonstrate that bromophenols 2,3,6-tribromo-4,5-dihydroxybenzyl alcohol (1), 2,3,6-tribromo-4,5-dihydroxybenzyl methyl ether (2), and bis-(2,3,6-tribromo-4,5-dihydroxybenzyl) ether (3) from red alga Symphyocladia latiuscula are moderate-selective human monoamine oxidase-A inhibitors and good dopamine D3/D4 receptor agonists. Bromophenol 3 showed a promising D4R agonist effect with a low micromole 50% effective concentration (EC50) value. All of the test ligands were docked against a three-dimensional (3D) model of hD3R and hD4R, and the result demonstrated strong binding through interaction with prime interacting residues-Asp110, Cys114, and His349 on hD3R and Asp115 and Cys119 on hD4R. Overall, the results demonstrated natural bromophenols, especially 1 and 3, from Symphyocladia latiuscula as multitarget ligands for neuroprotection, especially in PD and schizophrenia.
Collapse
Affiliation(s)
- Pradeep Paudel
- Department of Food and Life Science , Pukyong National University , Busan 48513 , Republic of Korea
| | - Se Eun Park
- Department of Food and Life Science , Pukyong National University , Busan 48513 , Republic of Korea
| | - Su Hui Seong
- Department of Food and Life Science , Pukyong National University , Busan 48513 , Republic of Korea
| | - Hyun Ah Jung
- Department of Food Science and Human Nutrition , Jeonbuk National University , Jeonju 54896 , Republic of Korea
| | - Jae Sue Choi
- Department of Food and Life Science , Pukyong National University , Busan 48513 , Republic of Korea
| |
Collapse
|
25
|
Yang P, Perlmutter JS, Benzinger TLS, Morris JC, Xu J. Dopamine D3 receptor: A neglected participant in Parkinson Disease pathogenesis and treatment? Ageing Res Rev 2020; 57:100994. [PMID: 31765822 PMCID: PMC6939386 DOI: 10.1016/j.arr.2019.100994] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/13/2019] [Accepted: 11/20/2019] [Indexed: 12/20/2022]
Abstract
Parkinson disease (PD) is a neurodegenerative disorder characterized by motor and non-motor symptoms which relentlessly and progressively lead to substantial disability and economic burden. Pathologically, these symptoms follow the loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc) associated with abnormal α-synuclein (α-Syn) deposition as cytoplasmic inclusions called Lewy bodies in pigmented brainstem nuclei, and in dystrophic neurons in striatal and cortical regions (Lewy neurites). Pharmacotherapy for PD focuses on improving quality of life and primarily targets dopaminergic pathways. Dopamine acts through two families of receptors, dopamine D1-like and dopamine D2-like; dopamine D3 receptors (D3R) belong to dopamine D2 receptor (D2R) family. Although D3R's precise role in the pathophysiology and treatment of PD has not been determined, we present evidence suggesting an important role for D3R in the early development and occurrence of PD. Agonist activation of D3R increases dopamine concentration, decreases α-Syn accumulation, enhances secretion of brain derived neurotrophic factors (BDNF), ameliorates neuroinflammation, alleviates oxidative stress, promotes neurogenesis in the nigrostriatal pathway, interacts with D1R to reduce PD associated motor symptoms and ameliorates side effects of levodopa (L-DOPA) treatment. Furthermore, D3R mutations can predict PD age of onset and prognosis of PD treatment. The role of D3R in PD merits further research. This review elucidates the potential role of D3R in PD pathogenesis and therapy.
Collapse
Affiliation(s)
- Pengfei Yang
- Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Blvd, St. Louis, MO 63110, USA
| | - Joel S Perlmutter
- Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Blvd, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, 510 S. Kingshighway Blvd, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University School of Medicine, 510 S. Kingshighway Blvd, St. Louis, MO 63110, USA; Department of Physical Therapy, Washington University School of Medicine, 510 S. Kingshighway Blvd, St. Louis, MO 63110, USA; Department of Occupational Therapy, Washington University School of Medicine, 510 S. Kingshighway Blvd, St. Louis, MO 63110, USA
| | - Tammie L S Benzinger
- Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Blvd, St. Louis, MO 63110, USA
| | - John C Morris
- Department of Neurology, Washington University School of Medicine, 510 S. Kingshighway Blvd, St. Louis, MO 63110, USA
| | - Jinbin Xu
- Department of Radiology, Washington University School of Medicine, 510 S. Kingshighway Blvd, St. Louis, MO 63110, USA.
| |
Collapse
|
26
|
Lanza K, Chemakin K, Lefkowitz S, Saito C, Chambers N, Bishop C. Reciprocal cross-sensitization of D1 and D3 receptors following pharmacological stimulation in the hemiparkinsonian rat. Psychopharmacology (Berl) 2020; 237:155-165. [PMID: 31435690 DOI: 10.1007/s00213-019-05353-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 08/15/2019] [Indexed: 12/20/2022]
Abstract
In the majority of Parkinson's disease (PD) patients, long-term dopamine (DA) replacement therapy leads to dyskinesia characterized by abnormal involuntary movements (AIMs). There are various mechanisms of dyskinesia, such as the sensitization of striatal DA D1 receptors (D1R) and upregulation of DA D3 receptors (D3R). These receptors interact physically and functionally in D1R-bearing medium spiny neurons to synergistically drive dyskinesia. However, the cross-receptor-mediated effects due to D1R-D3R cooperativity are still poorly understood. In pursuit of this, we examined whether or not pharmacological D1R or D3R stimulation sensitizes the dyskinetic response to the appositional agonist, a process known as cross-sensitization. First, we established D1R-D3R behavioral synergy in a cohort of 6-OHDA-lesioned female adult Sprague-Dawley rats. Then, in a new cohort, we tested for cross-sensitization in a between-subject design. Five groups received a sub-chronic regimen of either saline, the D1R agonist SKF38393 (1.0 mg/kg), or the D3R agonist PD128907 (0.3 mg/kg). For the final injection, each group received an acute injection of the other agonist. AIMs were monitored following each injection. Sub-chronic administration of both SKF38393 and PD128907 induced the development of dyskinesia. More importantly, cross-agonism tests revealed reciprocal cross-sensitization; chronic treatment with either SKF38393 or PD128907 induced sensitization to a single administration of the other agonist. This reciprocity was not marked by changes to either D1R or D3R striatal mRNA expression. The current study provides key behavioral data demonstrating the role of D3R in dyskinesia and provides behavioral evidence of D1R and D3R functional interactions.
Collapse
MESH Headings
- 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology
- Animals
- Benzopyrans/pharmacology
- Corpus Striatum/drug effects
- Corpus Striatum/metabolism
- Dopamine/metabolism
- Dopamine Agonists/pharmacology
- Dyskinesia, Drug-Induced/metabolism
- Female
- Oxazines/pharmacology
- Oxidopamine
- Parkinson Disease, Secondary/chemically induced
- Parkinson Disease, Secondary/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Dopamine D1/metabolism
- Receptors, Dopamine D3/metabolism
Collapse
Affiliation(s)
- Kathryn Lanza
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - Katherine Chemakin
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - Sarah Lefkowitz
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - Carolyn Saito
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - Nicole Chambers
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA
| | - Christopher Bishop
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, 4400 Vestal Parkway East, Binghamton, NY, 13902, USA.
| |
Collapse
|
27
|
Battiti FO, Cemaj SL, Guerrero AM, Shaik AB, Lam J, Rais R, Slusher BS, Deschamps JR, Imler GH, Newman AH, Bonifazi A. The Significance of Chirality in Drug Design and Synthesis of Bitopic Ligands as D 3 Receptor (D 3R) Selective Agonists. J Med Chem 2019; 62:6287-6314. [PMID: 31257877 DOI: 10.1021/acs.jmedchem.9b00702] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Because of the large degree of homology among dopamine D2-like receptors, discovering ligands capable of discriminating between the D2, D3, and D4 receptor subtypes remains a significant challenge. Previous work has exemplified the use of bitopic ligands as a powerful strategy in achieving subtype selectivity for agonists and antagonists alike. Inspired by the potential for chemical modification of the D3 preferential agonists (+)-PD128,907 (1) and PF592,379 (2), we synthesized bitopic structures to further improve their D3R selectivity. We found that the (2S,5S) conformation of scaffold 2 resulted in a privileged architecture with increased affinity and selectivity for the D3R. In addition, a cyclopropyl moiety incorporated into the linker and full resolution of the chiral centers resulted in lead compound 53 and eutomer 53a that demonstrate significantly higher D3R binding selectivities than the reference compounds. Moreover, the favorable metabolic stability in rat liver microsomes supports future studies in in vivo models of dopamine system dysregulation.
Collapse
Affiliation(s)
- Francisco O Battiti
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program , National Institutes of Health , 333 Cassell Drive , Baltimore , Maryland 21224 , United States
| | - Sophie L Cemaj
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program , National Institutes of Health , 333 Cassell Drive , Baltimore , Maryland 21224 , United States
| | - Adrian M Guerrero
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program , National Institutes of Health , 333 Cassell Drive , Baltimore , Maryland 21224 , United States
| | - Anver Basha Shaik
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program , National Institutes of Health , 333 Cassell Drive , Baltimore , Maryland 21224 , United States
| | - Jenny Lam
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program , National Institutes of Health , 333 Cassell Drive , Baltimore , Maryland 21224 , United States.,Johns Hopkins Drug Discovery Program , Johns Hopkins School of Medicine , 855 N. Wolfe Street , Baltimore , Maryland 21205 , United States
| | - Rana Rais
- Johns Hopkins Drug Discovery Program , Johns Hopkins School of Medicine , 855 N. Wolfe Street , Baltimore , Maryland 21205 , United States
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery Program , Johns Hopkins School of Medicine , 855 N. Wolfe Street , Baltimore , Maryland 21205 , United States
| | - Jeffery R Deschamps
- Naval Research Laboratory , Code 6910, 4555 Overlook Avenue , Washington, DC 20375 , United States
| | - Greg H Imler
- Naval Research Laboratory , Code 6910, 4555 Overlook Avenue , Washington, DC 20375 , United States
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program , National Institutes of Health , 333 Cassell Drive , Baltimore , Maryland 21224 , United States
| | - Alessandro Bonifazi
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse-Intramural Research Program , National Institutes of Health , 333 Cassell Drive , Baltimore , Maryland 21224 , United States
| |
Collapse
|
28
|
de Oliveira PG, Ramos MLS, Amaro AJ, Dias RA, Vieira SI. G i/o-Protein Coupled Receptors in the Aging Brain. Front Aging Neurosci 2019; 11:89. [PMID: 31105551 PMCID: PMC6492497 DOI: 10.3389/fnagi.2019.00089] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/03/2019] [Indexed: 12/18/2022] Open
Abstract
Cells translate extracellular signals to regulate processes such as differentiation, metabolism and proliferation, via transmembranar receptors. G protein-coupled receptors (GPCRs) belong to the largest family of transmembrane receptors, with over 800 members in the human species. Given the variety of key physiological functions regulated by GPCRs, these are main targets of existing drugs. During normal aging, alterations in the expression and activity of GPCRs have been observed. The central nervous system (CNS) is particularly affected by these alterations, which results in decreased brain functions, impaired neuroregeneration, and increased vulnerability to neuropathologies, such as Alzheimer's and Parkinson diseases. GPCRs signal via heterotrimeric G proteins, such as Go, the most abundant heterotrimeric G protein in CNS. We here review age-induced effects of GPCR signaling via the Gi/o subfamily at the CNS. During the aging process, a reduction in protein density is observed for almost half of the Gi/o-coupled GPCRs, particularly in age-vulnerable regions such as the frontal cortex, hippocampus, substantia nigra and striatum. Gi/o levels also tend to decrease with aging, particularly in regions such as the frontal cortex. Alterations in the expression and activity of GPCRs and coupled G proteins result from altered proteostasis, peroxidation of membranar lipids and age-associated neuronal degeneration and death, and have impact on aging hallmarks and age-related neuropathologies. Further, due to oligomerization of GPCRs at the membrane and their cooperative signaling, down-regulation of a specific Gi/o-coupled GPCR may affect signaling and drug targeting of other types/subtypes of GPCRs with which it dimerizes. Gi/o-coupled GPCRs receptorsomes are thus the focus of more effective therapeutic drugs aiming to prevent or revert the decline in brain functions and increased risk of neuropathologies at advanced ages.
Collapse
Affiliation(s)
- Patrícia G de Oliveira
- Department of Medical Sciences, Institute of Biomedicine (iBiMED) and The Discovery CTR, Universidade de Aveiro, Aveiro, Portugal
| | - Marta L S Ramos
- Department of Medical Sciences, Institute of Biomedicine (iBiMED) and The Discovery CTR, Universidade de Aveiro, Aveiro, Portugal
| | - António J Amaro
- School of Health Sciences (ESSUA), Universidade de Aveiro, Aveiro, Portugal
| | - Roberto A Dias
- Department of Medical Sciences, Institute of Biomedicine (iBiMED) and The Discovery CTR, Universidade de Aveiro, Aveiro, Portugal
| | - Sandra I Vieira
- Department of Medical Sciences, Institute of Biomedicine (iBiMED) and The Discovery CTR, Universidade de Aveiro, Aveiro, Portugal
| |
Collapse
|
29
|
Vita A, Minelli A, Barlati S, Deste G, Giacopuzzi E, Valsecchi P, Turrina C, Gennarelli M. Treatment-Resistant Schizophrenia: Genetic and Neuroimaging Correlates. Front Pharmacol 2019; 10:402. [PMID: 31040787 PMCID: PMC6476957 DOI: 10.3389/fphar.2019.00402] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 04/01/2019] [Indexed: 12/11/2022] Open
Abstract
Schizophrenia is a severe neuropsychiatric disorder that affects approximately 0.5–1% of the population. Response to antipsychotic therapy is highly variable, and it is not currently possible to predict those patients who will or will not respond to antipsychotic medication. Furthermore, a high percentage of patients, approximately 30%, are classified as treatment-resistant (treatment-resistant schizophrenia; TRS). TRS is defined as a non-response to at least two trials of antipsychotic medication of adequate dose and duration. These patients are usually treated with clozapine, the only evidence-based pharmacotherapy for TRS. However, clozapine is associated with severe adverse events. For these reasons, there is an increasing interest to identify better targets for drug development of new compounds and to establish better biomarkers for existing medications. The ability of antipsychotics to improve psychotic symptoms is dependent on their antagonist and reverse agonist activities at different neuroreceptors, and some genetic association studies of TRS have focused on different pharmacodynamic factors. Some genetic studies have shown an association between antipsychotic response or TRS and neurodevelopment candidate genes, antipsychotic mechanisms of action (such as dopaminergic, serotonergic, GABAergic, and glutamatergic) or pharmacokinetic factors (i.e., differences in the cytochrome families). Moreover, there is a growing body of literature on the structural and functional neuroimaging research into TRS. Neuroimaging studies can help to uncover the underlying neurobiological reasons for such resistance and identify resistant patients earlier. Studies examining the neuropharmacological mechanisms of antipsychotics, including clozapine, can help to improve our knowledge of their action on the central nervous system, with further implications for the discovery of biomarkers and the development of new treatments. The identification of the underlying mechanisms of TRS is a major challenge for developing personalized medicine in the psychiatric field for schizophrenia treatment. The main goal of precision medicine is to use genetic and brain-imaging information to improve the safety, effectiveness, and health outcomes of patients via more efficiently targeted risk stratification, prevention, and tailored medication and treatment management approaches. The aim of this review is to summarize the state of art of pharmacogenetic, pharmacogenomic and neuroimaging studies in TRS.
Collapse
Affiliation(s)
- Antonio Vita
- Department of Mental Health and Addiction Services, ASST Spedali Civili, Brescia, Italy.,Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alessandra Minelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Stefano Barlati
- Department of Mental Health and Addiction Services, ASST Spedali Civili, Brescia, Italy.,Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Giacomo Deste
- Department of Mental Health and Addiction Services, ASST Spedali Civili, Brescia, Italy
| | - Edoardo Giacopuzzi
- Genetic Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Paolo Valsecchi
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Cesare Turrina
- Department of Mental Health and Addiction Services, ASST Spedali Civili, Brescia, Italy.,Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Massimo Gennarelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.,Genetic Unit, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| |
Collapse
|
30
|
Liow JS, Morse CL, Lu S, Frankland M, Tye GL, Zoghbi SS, Gladding RL, Shaik AB, Innis RB, Newman AH, Pike VW. [ O- methyl- 11C] N-(4-(4-(3-Chloro-2-methoxyphenyl)-piperazin-1-yl)butyl)-1 H-indole-2-carboxamide ([ 11C]BAK4-51) Is an Efflux Transporter Substrate and Ineffective for PET Imaging of Brain D₃ Receptors in Rodents and Monkey. Molecules 2018; 23:molecules23112737. [PMID: 30360553 PMCID: PMC6278341 DOI: 10.3390/molecules23112737] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/15/2018] [Accepted: 10/18/2018] [Indexed: 11/16/2022] Open
Abstract
Selective high-affinity antagonists for the dopamine D₃ receptor (D₃R) are sought for treating substance use disorders. Positron emission tomography (PET) with an effective D₃R radioligand could be a useful tool for the development of such therapeutics by elucidating pharmacological specificity and target engagement in vivo. Currently, a D₃R-selective radioligand does not exist. The D₃R ligand, N-(4-(4-(3-chloro-2-methoxyphenyl)piperazin-1-yl)butyl)-1H-indole-2-carboxamide (BAK4-51, 1), has attractive properties for PET radioligand development, including full antagonist activity, very high D₃R affinity, D₃R selectivity, and moderate lipophilicity. We labeled 1 with the positron-emitter carbon-11 (t1/2 = 20.4 min) in the methoxy group for evaluation as a radioligand in animals with PET. However, [11C]1 was found to be an avid substrate for brain efflux transporters and lacked D₃R-specific signal in rodent and monkey brain in vivo.
Collapse
Affiliation(s)
- Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Room B3C346, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Room B3C346, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Shuiyu Lu
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Room B3C346, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Michael Frankland
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Room B3C346, 10 Center Drive, Bethesda, MD 20892, USA.
| | - George L Tye
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Room B3C346, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Room B3C346, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Robert L Gladding
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Room B3C346, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Anver B Shaik
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, USA.
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Room B3C346, 10 Center Drive, Bethesda, MD 20892, USA.
| | - Amy H Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, 333 Cassell Drive, Baltimore, MD 21224, USA.
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Room B3C346, 10 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
31
|
Derouiche L, Massotte D. G protein-coupled receptor heteromers are key players in substance use disorder. Neurosci Biobehav Rev 2018; 106:73-90. [PMID: 30278192 DOI: 10.1016/j.neubiorev.2018.09.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 09/25/2018] [Accepted: 09/26/2018] [Indexed: 12/19/2022]
Abstract
G protein-coupled receptors (GPCR) represent the largest family of membrane proteins in the human genome. Physical association between two different GPCRs is linked to functional interactions which generates a novel entity, called heteromer, with specific ligand binding and signaling properties. Heteromerization is increasingly recognized to take place in the mesocorticolimbic pathway and to contribute to various aspects related to substance use disorder. This review focuses on heteromers identified in brain areas relevant to drug addiction. We report changes at the molecular and cellular levels that establish specific functional impact and highlight behavioral outcome in preclinical models. Finally, we briefly discuss selective targeting of native heteromers as an innovative therapeutic option.
Collapse
Affiliation(s)
- Lyes Derouiche
- Institut des Neurosciences Cellulaires et Integratives, UPR 3212, 5 rue Blaise Pascal, F-67000 Strasbourg, France
| | - Dominique Massotte
- Institut des Neurosciences Cellulaires et Integratives, UPR 3212, 5 rue Blaise Pascal, F-67000 Strasbourg, France.
| |
Collapse
|
32
|
Zhan J, Jordan CJ, Bi GH, He XH, Gardner EL, Wang YL, Xi ZX. Genetic deletion of the dopamine D3 receptor increases vulnerability to heroin in mice. Neuropharmacology 2018; 141:11-20. [PMID: 30138692 DOI: 10.1016/j.neuropharm.2018.08.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 07/31/2018] [Accepted: 08/17/2018] [Indexed: 12/21/2022]
Abstract
Despite extensive research, the neurobiological risk factors that convey vulnerability to opioid abuse are still unknown. Recent studies suggest that the dopamine D3 receptor (D3R) is involved in opioid self-administration, but it remains unclear whether altered D3R availability is a risk factor for the development of opioid abuse and addiction. Here we used dopamine D3 receptor-knockout (D3-KO) mice to investigate the role of this receptor in the different phases of opioid addiction. D3-KO mice learned to self-administer heroin faster and took more heroin than wild-type mice during acquisition and maintenance of self-administration. D3R-KO mice also displayed higher motivation to work to obtain heroin reward during self-administration under progressive-ratio reinforcement, as well as elevated heroin-seeking during extinction and reinstatement testing. In addition, deletion of the D3R induced higher baseline levels of extracellular dopamine (DA) in the nucleus accumbens (NAc), higher basal levels of locomotion, and reduced NAc DA and locomotor responses to lower doses of heroin. These findings suggest that the D3R is critically involved in regulatory processes that normally limit opioid intake via DA-related mechanisms. Deletion of D3R augments opioid-taking and opioid-seeking behaviors. Therefore, low D3R availability in the brain may represent a risk factor for the development of opioid abuse and addiction.
Collapse
Affiliation(s)
- Jia Zhan
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA; Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Chloe J Jordan
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Guo-Hua Bi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Xiang-Hu He
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA; Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Eliot L Gardner
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA
| | - Yan-Lin Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China.
| | - Zheng-Xiong Xi
- Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD, 21224, USA.
| |
Collapse
|