1
|
Tournier BB, Ceyzériat K, Badina AM, Gloria Y, Fall AB, Amossé Q, Tsartsalis S, Millet P. Impairment of hippocampal astrocyte-mediated striatal dopamine release and locomotion in Alzheimer's disease. Neuroimage 2024; 298:120778. [PMID: 39122057 DOI: 10.1016/j.neuroimage.2024.120778] [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: 01/08/2024] [Revised: 07/01/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Clinical and translational research has identified deficits in the dopaminergic neurotransmission in the striatum in Alzheimer's disease (AD) and this could be related to the pathophysiology of psychiatric symptoms appearing even at early stages of the pathology. HYPOTHESIS We hypothesized that AD pathology in the hippocampus may influence dopaminergic neurotransmission even in the absence of AD-related lesion in the mesostriatal circuit. METHODS We chemogenetically manipulated the activity of hippocampal neurons and astrocytes in wild-type and hemizygous TgF344-AD (Tg) rats, an animal model of AD pathology. We assessed the brain-wide functional output of this manipulation using in vivo Single Photon Emission Computed Tomography to measure cerebral blood flow and D2/3 receptor binding, in response to acute (3 mg kg-1 i.p.) and chronic (0.015 mg/ml in drinking water, 28 days) stimulation of neurons or astrocytes with clozapine N-oxide. We also assessed the effects of the chronic chemogenetic manipulations on D2 receptor density, low or high aggregated forms of amyloid Aβ40 and Aβ42, astrocytes and microglial reactivity, and the capacity of astrocytes and microglia to surround and phagocytize Aβ both locally and in the striatum. RESULTS We showed that acute and chronic neuronal and astrocytic stimulation induces widespread effects on the brain regional activation pattern, notably with an inhibition of striatal activation. In the Tg rats, both these effects were blunted. Chemogenetic stimulation in the hippocampus increased microglial density and its capacity to limit AD pathology, whereas these effects were absent in the striatum perhaps as a consequence of the altered connectivity between the hippocampus and the striatum. CONCLUSIONS Our work suggests that hippocampal AD pathology may alter mesostriatal signalling and induce widespread alterations of brain activity. Neuronal and astrocytic activation may induce a protective, Aβ-limiting phenotype of microglia, which surrounds Aβ plaques and limits Αβ concentration more efficiently.
Collapse
Affiliation(s)
- Benjamin B Tournier
- Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland; Department of Psychiatry, University of Geneva, Geneva, Switzerland.
| | - Kelly Ceyzériat
- Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland; Division of Nuclear medicine and Molecular Imaging, University Hospitals of Geneva, Geneva, Switzerland; Division of Radiation Oncology, Department of Oncology, University Hospitals of Geneva, Geneva, Switzerland
| | | | - Yesica Gloria
- Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland; Department of Psychiatry, University of Geneva, Geneva, Switzerland; Present address: Bertarelli Foundation Gene Therapy Platform, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Aïda B Fall
- Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland; Division of Geriatric Psychiatry, Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Quentin Amossé
- Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland; Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Stergios Tsartsalis
- Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland; Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Philippe Millet
- Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland; Department of Psychiatry, University of Geneva, Geneva, Switzerland
| |
Collapse
|
2
|
Tang Y, Li Y, Cao P, Dong Y, Xu G, Si Q, Li R, Sui Y. Striatum and globus pallidus structural abnormalities in schizophrenia: A retrospective study of the different stages of the disease. Prog Neuropsychopharmacol Biol Psychiatry 2024; 133:111022. [PMID: 38692473 DOI: 10.1016/j.pnpbp.2024.111022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/14/2024] [Accepted: 04/28/2024] [Indexed: 05/03/2024]
Abstract
BACKGROUND The basal ganglia are important structures for the release of dopamine in the limbic circuits of the midbrain, and the striatum and globus pallidus are the major nuclei of the basal ganglia, and the dysfunction of these regions has been the basis of many models that have attempted to explain the underlying mechanisms of schizophrenia symptoms. The purpose of this study was to investigate the changes in the volume of the striatum subregion and globus pallidus in three different stages of schizophrenia, and to analyze whether these volume changes were related to antipsychotic drugs and schizophrenia symptoms. METHODS In this study, we investigated the volume of the striatum and globus pallidus in patients with schizophrenia at three different stages. The study included 57 patients with first-episode schizophrenia (FSZ), 51 patients with early-stage schizophrenia (ESZ), 86 patients with chronic schizophrenia (CSZ), and 191 healthy controls (HC), all of whom underwent structured magnetic resonance imaging (MRI) scans. Covariance analysis was performed using SPSS 26.0 was used for covariance analysis to determine whether there were significant differences in striatal subregion and globus pallidus volume between groups, and stratified analysis was used to further eliminate the effect of age on brain volume. Finally, the correlation analysis between the region of interest and the cumulative dose of antipsychotic drugs and psychotic symptoms was performed. RESULTS The comparison between the different stages of the illness showed significant volume differences in the left caudate nucleus (lCAU) (F = 2.665, adjusted p = 0.048), left putamen (lPUT) (F = 12.749, adjusted p < 0.001), left pallidum (lPAL) (F = 41.111, adjusted p < 0.001), and right pallidum (rPAL) (F = 14.479, adjusted p < 0.001). Post-hoc analysis with corrections showed that the volume differences in the lCAU subregion disappeared. Further stratified analysis controlling for age showed that compared with the HC, the lPAL (t = 4.347, p < 0.001) was initially significantly enlarged in the FSZ group, the lPUT (t = 4.493, p < 0.001), rPUT (t = 2.190, p = 0.031), lPAL (t = 7.894, p < 0.001), and rPAL (t = 4.983, p < 0.001) volumes were all significantly increased in the ESZ group, and the lPUT (t = 3.314, p = 0.002), lPAL (t = 6.334, p < 0.001), and rPAL (t = 3.604, p < 0.001) subregion volumes were also significantly increased in the CSZ group. Correlation analysis showed that lPUT and bilateral globus pallidus were associated with cumulative dose of antipsychotics, but were not associated with clinical symptoms in each subregion. CONCLUSION The findings suggest that different subregions of the striatum and globus pallidus show significant volume differences at different stages of schizophrenia compared to HC. These volume differences may be strong radiographic evidence for schizophrenia. In addition, the lPAL was the only significantly different brain region observed in the FSZ group, suggesting that it may be a sensitive indicator of early brain structural changes in schizophrenia. Finally, our findings support the hypothesis that antipsychotic drugs have an effect on the volume of brain structures.
Collapse
Affiliation(s)
- Yilin Tang
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, China
| | - Yuting Li
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, China
| | - Peiyu Cao
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, China
| | - Yingbo Dong
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, China
| | - Guoxin Xu
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, China
| | - Qi Si
- Huai'an No. 3 People's Hospital, China
| | | | - Yuxiu Sui
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, China.
| |
Collapse
|
3
|
Darcey VL, Guo J, Chi M, Chung ST, Courville AB, Gallagher I, Herscovitch P, Howard R, LaNoire M, Milley L, Schick A, Stagliano M, Turner S, Urbanski N, Yang S, Yim E, Zhai N, Zhou MS, Hall KD. Striatal dopamine tone is positively associated with body mass index in humans as determined by PET using dual dopamine type-2 receptor antagonist tracers. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.27.23296169. [PMID: 37886556 PMCID: PMC10602123 DOI: 10.1101/2023.09.27.23296169] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
The relationship between adiposity and dopamine type-2 receptor binding potential (D2BP) in the human brain has been repeatedly studied for >20 years with highly discrepant results, likely due to variable methodologies and differing study populations. We conducted a controlled inpatient feeding study to measure D2BP in the striatum using positron emission tomography with both [18F]fallypride and [11C]raclopride in pseudo-random order in 54 young adults with a wide range of body mass index (BMI 20-44 kg/m2). Within-subject D2BP measurements using the two tracers were moderately correlated (r=0.47, p<0.001). D2BP was negatively correlated with BMI as measured by [11C]raclopride (r= -0.51; p<0.0001) but not [18F]fallypride (r=-0.01; p=0.92) and these correlation coefficients were significantly different from each other (p<0.001). Given that [18F]fallypride has greater binding affinity to dopamine type-2 receptors than [11C]raclopride, which is more easily displaced by endogenous dopamine, our results suggest that adiposity is positively associated with increased striatal dopamine tone.
Collapse
Affiliation(s)
- Valerie L Darcey
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
- Center on Compulsive Behaviors, Intramural Research Program, NIH, Bethesda, MD, USA
| | - Juen Guo
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Meible Chi
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stephanie T Chung
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Amber B Courville
- Human Energy and Body Weight Regulation Core, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Isabelle Gallagher
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Peter Herscovitch
- Positron Emission Tomography Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Rebecca Howard
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Melissa LaNoire
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lauren Milley
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alex Schick
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael Stagliano
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sara Turner
- Nutrition Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Nicholas Urbanski
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shanna Yang
- Nutrition Department, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Eunha Yim
- University of Maryland, College Park, MD, USA
| | - Nan Zhai
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Megan S Zhou
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kevin D Hall
- Integrative Physiology Section, National Institute of Diabetes & Digestive & Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
4
|
Smigielski L, Wotruba D, Treyer V, Rössler J, Papiol S, Falkai P, Grünblatt E, Walitza S, Rössler W. The Interplay Between Postsynaptic Striatal D2/3 Receptor Availability, Adversity Exposure and Odd Beliefs: A [11C]-Raclopride PET Study. Schizophr Bull 2021; 47:1495-1508. [PMID: 33876249 PMCID: PMC8379534 DOI: 10.1093/schbul/sbab034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Between unaffected mental health and diagnosable psychiatric disorders, there is a vast continuum of functioning. The hypothesized link between striatal dopamine signaling and psychosis has guided a prolific body of research. However, it has been understudied in the context of multiple interacting factors, subclinical phenotypes, and pre-postsynaptic dynamics. METHOD This work investigated psychotic-like experiences and D2/3 dopamine postsynaptic receptor availability in the dorsal striatum, quantified by in vivo [11C]-raclopride positron emission tomography, in a sample of 24 healthy male individuals. Additional mediation and moderation effects with childhood trauma and key dopamine-regulating genes were examined. RESULTS An inverse relationship between nondisplaceable binding potential and subclinical symptoms was identified. D2/3 receptor availability in the left putamen fully mediated the association between traumatic childhood experiences and odd beliefs, that is, inclinations to see meaning in randomness and unfounded interpretations. Moreover, the effect of early adversity was moderated by a DRD2 functional variant (rs1076560). The results link environmental and neurobiological influences in the striatum to the origination of psychosis spectrum symptomology, consistent with the social defeat and diathesis-stress models. CONCLUSIONS Adversity exposure may affect the dopamine system as in association with biases in probabilistic reasoning, attributional style, and salience processing. The inverse relationship between D2/3 availability and symptomology may be explained by endogenous dopamine occupying the receptor, postsynaptic compensatory mechanisms, and/or altered receptor sensitivity. This may also reflect a cognitively stabilizing mechanism in non-help-seeking individuals. Future research should comprehensively characterize molecular parameters of dopamine neurotransmission along the psychosis spectrum and according to subtype profiling.
Collapse
Affiliation(s)
- Lukasz Smigielski
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, Zurich Program for Sustainable Development of Mental Health Services (ZInEP), University of Zurich, Zurich, Switzerland,Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland,To whom correspondence should be addressed; Psychiatric University Hospital Zurich, Militärstrasse 8, 8004 Zurich, Switzerland; tel: +044-296-73-94, fax: +044-296-74-69, e-mail:
| | - Diana Wotruba
- Collegium Helveticum, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Valerie Treyer
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland,Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
| | - Julian Rössler
- Institute of Anesthesiology, University Hospital Zurich, Zurich, Switzerland
| | - Sergi Papiol
- Institute of Psychiatric Phenomics and Genomics, University Hospital, Ludwig Maximilian University, Munich, Germany,Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Edna Grünblatt
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Susanne Walitza
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland,Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Wulf Rössler
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric University Hospital Zurich, Zurich Program for Sustainable Development of Mental Health Services (ZInEP), University of Zurich, Zurich, Switzerland,Department of Psychiatry and Psychotherapy, Charité Universitätsmedizin, Campus Charité Mitte, Berlin, Germany,Laboratory of Neuroscience (LIM 27), Institute of Psychiatry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
5
|
Goud NS, Bhattacharya A, Joshi RK, Nagaraj C, Bharath RD, Kumar P. Carbon-11: Radiochemistry and Target-Based PET Molecular Imaging Applications in Oncology, Cardiology, and Neurology. J Med Chem 2021; 64:1223-1259. [PMID: 33499603 DOI: 10.1021/acs.jmedchem.0c01053] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The positron emission tomography (PET) molecular imaging technique has gained its universal value as a remarkable tool for medical diagnosis and biomedical research. Carbon-11 is one of the promising radiotracers that can report target-specific information related to its pharmacology and physiology to understand the disease status. Currently, many of the available carbon-11 (t1/2 = 20.4 min) PET radiotracers are heterocyclic derivatives that have been synthesized using carbon-11 inserted different functional groups obtained from primary and secondary carbon-11 precursors. A spectrum of carbon-11 PET radiotracers has been developed against many of the upregulated and emerging targets for the diagnosis, prognosis, prediction, and therapy in the fields of oncology, cardiology, and neurology. This review focuses on the carbon-11 radiochemistry and various target-specific PET molecular imaging agents used in tumor, heart, brain, and neuroinflammatory disease imaging along with its associated pathology.
Collapse
Affiliation(s)
- Nerella Sridhar Goud
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Ahana Bhattacharya
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Raman Kumar Joshi
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Chandana Nagaraj
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Rose Dawn Bharath
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Pardeep Kumar
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| |
Collapse
|
6
|
Tronchin G, Akudjedu TN, Ahmed M, Holleran L, Hallahan B, Cannon DM, McDonald C. Progressive subcortical volume loss in treatment-resistant schizophrenia patients after commencing clozapine treatment. Neuropsychopharmacology 2020; 45:1353-1361. [PMID: 32268345 PMCID: PMC7298040 DOI: 10.1038/s41386-020-0665-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 02/07/2023]
Abstract
The association of antipsychotic medication with abnormal brain morphometry in schizophrenia remains uncertain. This study investigated subcortical morphometric changes 6 months after switching treatment to clozapine in patients with treatment-resistant schizophrenia compared with healthy volunteers, and the relationships between longitudinal volume changes and clinical variables. In total, 1.5T MRI images were acquired at baseline before commencing clozapine and again after 6 months of treatment for 33 patients with treatment-resistant schizophrenia and 31 controls, and processed using the longitudinal pipeline of Freesurfer v.5.3.0. Two-way repeated MANCOVA was used to assess group differences in subcortical volumes over time and partial correlations to determine association with clinical variables. Whereas no significant subcortical volume differences were found between patients and controls at baseline (F(8,52) = 1.79; p = 0.101), there was a significant interaction between time, group and structure (F(7,143) = 52.54; p < 0.001). Corrected post-hoc analyses demonstrated that patients had significant enlargement of lateral ventricles (F(1,59) = 48.89; p < 0.001) and reduction of thalamus (F(1,59) = 34.85; p < 0.001), caudate (F(1,59) = 59.35; p < 0.001), putamen (F(1,59) = 87.20; p < 0.001) and hippocampus (F(1,59) = 14.49; p < 0.001) volumes. Thalamus and putamen volume reduction was associated with improvement in PANSS (r = 0.42; p = 0.021, r = 0.39; p = 0.033), SANS (r = 0.36; p = 0.049, r = 0.40; p = 0.027) and GAF (r = -0.39; p = 0.038, r = -0.42; p = 0.024) scores. Reduced thalamic volume over time was associated with increased serum clozapine level at follow-up (r = -0.44; p = 0.010). Patients with treatment-resistant schizophrenia display progressive subcortical volume deficits after switching to clozapine despite experiencing symptomatic improvement. Thalamo-striatal progressive volumetric deficit associated with symptomatic improvement after clozapine exposure may reflect an adaptive response related to improved outcome rather than a harmful process.
Collapse
Affiliation(s)
- Giulia Tronchin
- Centre for Neuroimaging & Cognitive Genomics (NICOG), Clinical Neuroimaging Laboratory, NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, H91TK33, Ireland.
| | - Theophilus N Akudjedu
- Centre for Neuroimaging & Cognitive Genomics (NICOG), Clinical Neuroimaging Laboratory, NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, H91TK33, Ireland
- Institute of Medical Imaging & Visualisation, Faculty of Health & Social Science, Department of Medical Science & Public Science, Bournemouth University, Bournemouth, UK
| | - Mohamed Ahmed
- Centre for Neuroimaging & Cognitive Genomics (NICOG), Clinical Neuroimaging Laboratory, NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, H91TK33, Ireland
| | - Laurena Holleran
- Centre for Neuroimaging & Cognitive Genomics (NICOG), Clinical Neuroimaging Laboratory, NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, H91TK33, Ireland
| | - Brian Hallahan
- Centre for Neuroimaging & Cognitive Genomics (NICOG), Clinical Neuroimaging Laboratory, NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, H91TK33, Ireland
| | - Dara M Cannon
- Centre for Neuroimaging & Cognitive Genomics (NICOG), Clinical Neuroimaging Laboratory, NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, H91TK33, Ireland
| | - Colm McDonald
- Centre for Neuroimaging & Cognitive Genomics (NICOG), Clinical Neuroimaging Laboratory, NCBES Galway Neuroscience Centre, College of Medicine Nursing and Health Sciences, National University of Ireland Galway, Galway, H91TK33, Ireland
| |
Collapse
|
7
|
Lu W, Guo W, Hou K, Zhao H, Shi L, Dong K, Qiu J. Grey matter differences associated with age and sex hormone levels between premenopausal and perimenopausal women: A voxel-based morphometry study. J Neuroendocrinol 2018; 30:e12655. [PMID: 30372794 DOI: 10.1111/jne.12655] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 09/22/2018] [Accepted: 10/24/2018] [Indexed: 11/30/2022]
Abstract
The present study aimed to explore brain morphological alterations associated with age and sex hormone levels between premenopausal and perimenopausal women using magnetic resonance imaging (MRI) T1 -weighted structural images. Thirty-two premenopausal women aged (mean ± SD) 47.75 ± 1.55 years and twenty-five recently perimenopausal women aged 51.60 ± 1.63 years were evaluated for sex hormone levels, including prolactin, follicle-stimulating hormone, luteinising hormone, oestradiol, free testosterone and progesterone. A 3.0-Tesla MRI scanner was utilised to acquire T1 images. Voxel-based morphometry (VBM) was used to evaluate changes in grey matter volume between the two groups. The general linear model was applied with false discovery rate correction for between group voxel-wise statistics. Spearman partial correlation analyses were conducted between age, sex hormone levels and regions of grey matter volume showing significant differences between the two groups. The VBM analysis revealed that age and menopause per se lead to grey matter volume reduction in certain brain structures. These structural changes might be potential causes of sexual dysfunction, nervous system degeneration and depression, which need to be examined in future studies. Our findings might provide evidence and guide future research in understanding the menopausal transition.
Collapse
Affiliation(s)
- Weizhao Lu
- Medical Engineering and Technical Center, Taishan Medcial University, Taian, China
- Department of Radiology, Taishan Medical University, Taian, China
| | - Wei Guo
- Affiliated Hospital of Taishan Medical University, Taian, China
| | - Kun Hou
- Medical Engineering and Technical Center, Taishan Medcial University, Taian, China
- Department of Radiology, Taishan Medical University, Taian, China
| | - Huihui Zhao
- Medical Engineering and Technical Center, Taishan Medcial University, Taian, China
- Department of Radiology, Taishan Medical University, Taian, China
| | - Liting Shi
- Medical Engineering and Technical Center, Taishan Medcial University, Taian, China
- Department of Radiology, Taishan Medical University, Taian, China
| | - Kejiang Dong
- Department of Radiology, Taishan Medical University, Taian, China
| | - Jianfeng Qiu
- Medical Engineering and Technical Center, Taishan Medcial University, Taian, China
- Department of Radiology, Taishan Medical University, Taian, China
| |
Collapse
|
8
|
van Galen KA, Ter Horst KW, Booij J, la Fleur SE, Serlie MJ. The role of central dopamine and serotonin in human obesity: lessons learned from molecular neuroimaging studies. Metabolism 2018; 85:325-339. [PMID: 28970033 DOI: 10.1016/j.metabol.2017.09.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/14/2017] [Accepted: 09/18/2017] [Indexed: 12/11/2022]
Abstract
Obesity results from an imbalance between energy intake and expenditure, and many studies have aimed to determine why obese individuals continue to (over)consume food under conditions of caloric excess. The two major "neurotransmitter hypotheses" of obesity state that increased food intake is partially driven by decreased dopamine-mediated reward and decreased serotonin-mediated homeostatic feedback in response to food intake. Using molecular neuroimaging studies to visualize and quantify aspects of the central dopamine and serotonin systems in vivo, recent PET and SPECT studies have also implicated alterations in these systems in human obesity. The interpretation of these data, however, is more complex than it may appear. Here, we discuss important characteristics and limitations of current radiotracer methods and use this framework to comprehensively review the available human data on central dopamine and serotonin in obesity. On the basis of the available evidence, we conclude that obesity is associated with decreased central dopaminergic and serotonergic signaling and that future research, especially in long-term follow-up and interventional settings, is needed to advance our understanding of the neuronal pathophysiology of obesity in humans.
Collapse
Affiliation(s)
- Katy A van Galen
- Department of Endocrinology and Metabolism, Academic Medical Center, Amsterdam, Netherlands
| | - Kasper W Ter Horst
- Department of Endocrinology and Metabolism, Academic Medical Center, Amsterdam, Netherlands
| | - Jan Booij
- Department of Nuclear Medicine, Academic Medical Center, Amsterdam, Netherlands
| | - Susanne E la Fleur
- Department of Endocrinology and Metabolism, Academic Medical Center, Amsterdam, Netherlands; Laboratory of Endocrinology, Department of Clinical Chemistry, Academic Medical Center, Amsterdam, Netherlands; Metabolism and Reward Group, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Mireille J Serlie
- Department of Endocrinology and Metabolism, Academic Medical Center, Amsterdam, Netherlands.
| |
Collapse
|
9
|
Aringhieri S, Carli M, Kolachalam S, Verdesca V, Cini E, Rossi M, McCormick PJ, Corsini GU, Maggio R, Scarselli M. Molecular targets of atypical antipsychotics: From mechanism of action to clinical differences. Pharmacol Ther 2018; 192:20-41. [PMID: 29953902 DOI: 10.1016/j.pharmthera.2018.06.012] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The introduction of atypical antipsychotics (AAPs) since the discovery of its prototypical drug clozapine has been a revolutionary pharmacological step for treating psychotic patients as these allow a significant recovery not only in terms of hospitalization and reduction in symptoms severity, but also in terms of safety, socialization and better rehabilitation in the society. Regarding the mechanism of action, AAPs are weak D2 receptor antagonists and they act beyond D2 antagonism, involving other receptor targets which regulate dopamine and other neurotransmitters. Consequently, AAPs present a significant reduction of deleterious side effects like parkinsonism, hyperprolactinemia, apathy and anhedonia, which are all linked to the strong blockade of D2 receptors. This review revisits previous and current findings within the class of AAPs and highlights the differences in terms of receptor properties and clinical activities among them. Furthermore, we propose a continuum spectrum of "atypia" that begins with risperidone (the least atypical) to clozapine (the most atypical), while all the other AAPs fall within the extremes of this spectrum. Clozapine is still considered the gold standard in refractory schizophrenia and in psychoses present in Parkinson's disease, though it has been associated with adverse effects like agranulocytosis (0.7%) and weight gain, pushing the scientific community to find new drugs as effective as clozapine, but devoid of its side effects. To achieve this, it is therefore imperative to characterize and compare in depth the very complex molecular profile of AAPs. We also introduce relatively new concepts like biased agonism, receptor dimerization and neurogenesis to identify better the old and new hallmarks of "atypia". Finally, a detailed confrontation of clinical differences among the AAPs is presented, especially in relation to their molecular targets, and new means like therapeutic drug monitoring are also proposed to improve the effectiveness of AAPs in clinical practice.
Collapse
Affiliation(s)
- Stefano Aringhieri
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Marco Carli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Shivakumar Kolachalam
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Valeria Verdesca
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Enrico Cini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Mario Rossi
- Institute of Molecular Cell and Systems Biology, University of Glasgow, UK
| | - Peter J McCormick
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK
| | - Giovanni U Corsini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Roberto Maggio
- Biotechnological and Applied Clinical Sciences Department, University of L'Aquila, Italy
| | - Marco Scarselli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy.
| |
Collapse
|
10
|
Wiers CE, Cabrera EA, Tomasi D, Wong CT, Demiral ŞB, Kim SW, Wang GJ, Volkow ND. Striatal Dopamine D2/D3 Receptor Availability Varies Across Smoking Status. Neuropsychopharmacology 2017; 42:2325-2332. [PMID: 28643800 PMCID: PMC5645737 DOI: 10.1038/npp.2017.131] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/07/2017] [Accepted: 06/11/2017] [Indexed: 12/18/2022]
Abstract
To assess how tobacco smoking status affects baseline dopamine D2/D3 (D2R) receptor availability and methylphenidate-induced dopamine (DA) release, we retrospectively analyzed D2R availability measures of 8 current smokers, 10 ex-smokers, and 18 nonsmokers who were scanned with positron emission tomography and [11C]raclopride, after administration of an injection of placebo or 0.5 mg/kg i.v. methylphenidate. There was a significant effect of smoking status on baseline striatal D2R availability; with current smokers showing lower striatal D2R availability compared with nonsmokers (caudate, putamen, and ventral striatum) and with ex-smokers (caudate and putamen). Baseline striatal D2R did not differ between nonsmokers and ex-smokers. The effect of smoking status on methylphenidate-induced DA release tended to be lower in smokers but the difference was not significant (p=0.08). For behavioral measures, current smokers showed significantly higher aggression scores compared with both nonsmokers and ex-smokers. These results suggest that with abstinence ex-smokers may recover from low striatal D2R availability and from increased behavioral aggression seen in active smokers. However, longitudinal studies are needed to assess this within abstaining smokers.
Collapse
Affiliation(s)
- Corinde E Wiers
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Elizabeth A Cabrera
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Dardo Tomasi
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Christopher T Wong
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Şükrü B Demiral
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Sung Won Kim
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Gene-Jack Wang
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
- National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
11
|
Caravaggio F, Ku Chung J, Plitman E, Boileau I, Gerretsen P, Kim J, Iwata Y, Patel R, Chakravarty MM, Remington G, Graff-Guerrero A. The relationship between subcortical brain volume and striatal dopamine D 2/3 receptor availability in healthy humans assessed with [ 11 C]-raclopride and [ 11 C]-(+)-PHNO PET. Hum Brain Mapp 2017; 38:5519-5534. [PMID: 28752565 DOI: 10.1002/hbm.23744] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/21/2017] [Accepted: 07/16/2017] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Abnormalities in dopamine (DA) and brain morphology are observed in several neuropsychiatric disorders. However, it is not fully understood how these abnormalities may relate to one another. For such in vivo findings to be used as biomarkers for neuropsychiatric disease, it must be understood how variability in DA relates to brain structure under healthy conditions. We explored how the availability of striatal DA D2/3 receptors (D2/3 R) is related to the volume of subcortical brain structures in a sample of healthy humans. Differences in D2/3 R availability measured with an antagonist radiotracer ([11 C]-raclopride) versus an agonist radiotracer ([11 C]-(+)-PHNO) were examined. METHODS Data from 62 subjects scanned with [11 C]-raclopride (mean age = 38.98 ± 14.45; 23 female) and 68 subjects scanned with [11 C]-(+)-PHNO (mean age = 38.54 ± 14.59; 25 female) were used. Subcortical volumes were extracted from T1-weighted images using the Multiple Automatically Generated Templates (MAGeT-Brain) algorithm. Partial correlations were used controlling for age, gender, and total brain volume. RESULTS For [11 C]-(+)-PHNO, ventral caudate volumes were positively correlated with BPND in the dorsal caudate and globus pallidus (GP). Ventral striatum (VS) volumes were positively correlated with BPND in the VS. With [11 C]-raclopride, BPND in the VS was negatively correlated with subiculum volume of the hippocampus. Moreover, BPND in the GP was negatively correlated with the volume of the lateral posterior nucleus of the thalamus. CONCLUSION Findings are purely exploratory and presented corrected and uncorrected for multiple comparisons. We hope they will help inform the interpretation of future PET studies where concurrent changes in D2/3 R and brain morphology are observed. Hum Brain Mapp 38:5519-5534, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Fernando Caravaggio
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada.,Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada
| | - Jun Ku Chung
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada
| | - Eric Plitman
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada
| | - Isabelle Boileau
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada.,Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada
| | - Philip Gerretsen
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada.,Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada
| | - Julia Kim
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada
| | - Yusuke Iwata
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada.,Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada
| | - Raihaan Patel
- Department of Biological & Biomedical Engineering, McGill University, Montreal, Quebec, H4H 1R3, Canada.,Cerebral Imaging Centre, Douglas Mental Health Institute, McGill University, Montreal, Quebec, H4H 1R3, Canada
| | - M Mallar Chakravarty
- Department of Biological & Biomedical Engineering, McGill University, Montreal, Quebec, H4H 1R3, Canada.,Cerebral Imaging Centre, Douglas Mental Health Institute, McGill University, Montreal, Quebec, H4H 1R3, Canada.,Department of Psychiatry, McGill University, Montreal, Quebec, H4H 1R3, Canada
| | - Gary Remington
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada.,Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada
| | - Ariel Graff-Guerrero
- Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario, M5T 1R8, Canada.,Department of Psychiatry, University of Toronto, 250 College Street, Toronto, Ontario, M5T 1R8, Canada
| |
Collapse
|
12
|
Kubota M, Nagashima T, Takano H, Kodaka F, Fujiwara H, Takahata K, Moriguchi S, Kimura Y, Higuchi M, Okubo Y, Takahashi H, Ito H, Suhara T. Affinity States of Striatal Dopamine D2 Receptors in Antipsychotic-Free Patients with Schizophrenia. Int J Neuropsychopharmacol 2017; 20:928-935. [PMID: 29016872 PMCID: PMC5737675 DOI: 10.1093/ijnp/pyx063] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 07/19/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Dopamine D2 receptors are reported to have high-affinity (D2High) and low-affinity (D2Low) states. Although an increased proportion of D2High has been demonstrated in animal models of schizophrenia, few clinical studies have investigated this alteration of D2High in schizophrenia in vivo. METHODS Eleven patients with schizophrenia, including 10 antipsychotic-naive and 1 antipsychotic-free individuals, and 17 healthy controls were investigated. Psychopathology was assessed by Positive and Negative Syndrome Scale, and a 5-factor model was used. Two radioligands, [11C]raclopride and [11C]MNPA, were employed to quantify total dopamine D2 receptor and D2High, respectively, in the striatum by measuring their binding potentials. Binding potential values of [11C]raclopride and [11C]MNPA and the binding potential ratio of [11C]MNPA to [11C]raclopride in the striatal subregions were statistically compared between the 2 diagnostic groups using multivariate analysis of covariance controlling for age, gender, and smoking. Correlations between binding potential and Positive and Negative Syndrome Scale scores were also examined. RESULTS Multivariate analysis of covariance demonstrated a significant effect of diagnosis (schizophrenia and control) on the binding potential ratio (P=.018), although the effects of diagnosis on binding potential values obtained with either [11C]raclopride or [11C]MNPA were nonsignificant. Posthoc test showed that the binding potential ratio was significantly higher in the putamen of patients (P=.017). The Positive and Negative Syndrome Scale "depressed" factor in patients was positively correlated with binding potential values of both ligands in the caudate. CONCLUSIONS The present study indicates the possibilities of: (1) a higher proportion of D2High in the putamen despite unaltered amounts of total dopamine D2 receptors; and (2) associations between depressive symptoms and amounts of caudate dopamine D2 receptors in patients with schizophrenia.
Collapse
Affiliation(s)
- Manabu Kubota
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Kubota, Nagashima, Takano, Kodaka, Fujiwara, Moriguchi, Kimura, Higuchi, Takahashi, Ito, and Suhara); Department of Psychiatry, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan (Dr Takano); Department of Psychiatry, Jikei University School of Medicine, Tokyo, Japan (Dr Kodaka); Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan (Dr Kimura); Department of Neuropsychiatry, Nippon Medical School, Tokyo, Japan (Dr Okubo); Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan (Dr Takahashi); Department of Radiology, School of Medicine, Fukushima Medical University, Fukushima, Japan (Dr Ito)
| | - Tomohisa Nagashima
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Kubota, Nagashima, Takano, Kodaka, Fujiwara, Moriguchi, Kimura, Higuchi, Takahashi, Ito, and Suhara); Department of Psychiatry, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan (Dr Takano); Department of Psychiatry, Jikei University School of Medicine, Tokyo, Japan (Dr Kodaka); Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan (Dr Kimura); Department of Neuropsychiatry, Nippon Medical School, Tokyo, Japan (Dr Okubo); Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan (Dr Takahashi); Department of Radiology, School of Medicine, Fukushima Medical University, Fukushima, Japan (Dr Ito)
| | - Harumasa Takano
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Kubota, Nagashima, Takano, Kodaka, Fujiwara, Moriguchi, Kimura, Higuchi, Takahashi, Ito, and Suhara); Department of Psychiatry, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan (Dr Takano); Department of Psychiatry, Jikei University School of Medicine, Tokyo, Japan (Dr Kodaka); Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan (Dr Kimura); Department of Neuropsychiatry, Nippon Medical School, Tokyo, Japan (Dr Okubo); Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan (Dr Takahashi); Department of Radiology, School of Medicine, Fukushima Medical University, Fukushima, Japan (Dr Ito)
| | - Fumitoshi Kodaka
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Kubota, Nagashima, Takano, Kodaka, Fujiwara, Moriguchi, Kimura, Higuchi, Takahashi, Ito, and Suhara); Department of Psychiatry, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan (Dr Takano); Department of Psychiatry, Jikei University School of Medicine, Tokyo, Japan (Dr Kodaka); Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan (Dr Kimura); Department of Neuropsychiatry, Nippon Medical School, Tokyo, Japan (Dr Okubo); Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan (Dr Takahashi); Department of Radiology, School of Medicine, Fukushima Medical University, Fukushima, Japan (Dr Ito)
| | - Hironobu Fujiwara
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Kubota, Nagashima, Takano, Kodaka, Fujiwara, Moriguchi, Kimura, Higuchi, Takahashi, Ito, and Suhara); Department of Psychiatry, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan (Dr Takano); Department of Psychiatry, Jikei University School of Medicine, Tokyo, Japan (Dr Kodaka); Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan (Dr Kimura); Department of Neuropsychiatry, Nippon Medical School, Tokyo, Japan (Dr Okubo); Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan (Dr Takahashi); Department of Radiology, School of Medicine, Fukushima Medical University, Fukushima, Japan (Dr Ito)
| | - Keisuke Takahata
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Kubota, Nagashima, Takano, Kodaka, Fujiwara, Moriguchi, Kimura, Higuchi, Takahashi, Ito, and Suhara); Department of Psychiatry, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan (Dr Takano); Department of Psychiatry, Jikei University School of Medicine, Tokyo, Japan (Dr Kodaka); Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan (Dr Kimura); Department of Neuropsychiatry, Nippon Medical School, Tokyo, Japan (Dr Okubo); Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan (Dr Takahashi); Department of Radiology, School of Medicine, Fukushima Medical University, Fukushima, Japan (Dr Ito)
| | - Sho Moriguchi
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Kubota, Nagashima, Takano, Kodaka, Fujiwara, Moriguchi, Kimura, Higuchi, Takahashi, Ito, and Suhara); Department of Psychiatry, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan (Dr Takano); Department of Psychiatry, Jikei University School of Medicine, Tokyo, Japan (Dr Kodaka); Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan (Dr Kimura); Department of Neuropsychiatry, Nippon Medical School, Tokyo, Japan (Dr Okubo); Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan (Dr Takahashi); Department of Radiology, School of Medicine, Fukushima Medical University, Fukushima, Japan (Dr Ito)
| | - Yasuyuki Kimura
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Kubota, Nagashima, Takano, Kodaka, Fujiwara, Moriguchi, Kimura, Higuchi, Takahashi, Ito, and Suhara); Department of Psychiatry, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan (Dr Takano); Department of Psychiatry, Jikei University School of Medicine, Tokyo, Japan (Dr Kodaka); Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan (Dr Kimura); Department of Neuropsychiatry, Nippon Medical School, Tokyo, Japan (Dr Okubo); Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan (Dr Takahashi); Department of Radiology, School of Medicine, Fukushima Medical University, Fukushima, Japan (Dr Ito)
| | - Makoto Higuchi
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Kubota, Nagashima, Takano, Kodaka, Fujiwara, Moriguchi, Kimura, Higuchi, Takahashi, Ito, and Suhara); Department of Psychiatry, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan (Dr Takano); Department of Psychiatry, Jikei University School of Medicine, Tokyo, Japan (Dr Kodaka); Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan (Dr Kimura); Department of Neuropsychiatry, Nippon Medical School, Tokyo, Japan (Dr Okubo); Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan (Dr Takahashi); Department of Radiology, School of Medicine, Fukushima Medical University, Fukushima, Japan (Dr Ito)
| | - Yoshiro Okubo
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Kubota, Nagashima, Takano, Kodaka, Fujiwara, Moriguchi, Kimura, Higuchi, Takahashi, Ito, and Suhara); Department of Psychiatry, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan (Dr Takano); Department of Psychiatry, Jikei University School of Medicine, Tokyo, Japan (Dr Kodaka); Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan (Dr Kimura); Department of Neuropsychiatry, Nippon Medical School, Tokyo, Japan (Dr Okubo); Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan (Dr Takahashi); Department of Radiology, School of Medicine, Fukushima Medical University, Fukushima, Japan (Dr Ito)
| | - Hidehiko Takahashi
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Kubota, Nagashima, Takano, Kodaka, Fujiwara, Moriguchi, Kimura, Higuchi, Takahashi, Ito, and Suhara); Department of Psychiatry, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan (Dr Takano); Department of Psychiatry, Jikei University School of Medicine, Tokyo, Japan (Dr Kodaka); Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan (Dr Kimura); Department of Neuropsychiatry, Nippon Medical School, Tokyo, Japan (Dr Okubo); Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan (Dr Takahashi); Department of Radiology, School of Medicine, Fukushima Medical University, Fukushima, Japan (Dr Ito)
| | - Hiroshi Ito
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Kubota, Nagashima, Takano, Kodaka, Fujiwara, Moriguchi, Kimura, Higuchi, Takahashi, Ito, and Suhara); Department of Psychiatry, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan (Dr Takano); Department of Psychiatry, Jikei University School of Medicine, Tokyo, Japan (Dr Kodaka); Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan (Dr Kimura); Department of Neuropsychiatry, Nippon Medical School, Tokyo, Japan (Dr Okubo); Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan (Dr Takahashi); Department of Radiology, School of Medicine, Fukushima Medical University, Fukushima, Japan (Dr Ito)
| | - Tetsuya Suhara
- Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan (Drs Kubota, Nagashima, Takano, Kodaka, Fujiwara, Moriguchi, Kimura, Higuchi, Takahashi, Ito, and Suhara); Department of Psychiatry, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan (Dr Takano); Department of Psychiatry, Jikei University School of Medicine, Tokyo, Japan (Dr Kodaka); Department of Clinical and Experimental Neuroimaging, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, Obu, Japan (Dr Kimura); Department of Neuropsychiatry, Nippon Medical School, Tokyo, Japan (Dr Okubo); Department of Psychiatry, Graduate School of Medicine, Kyoto University, Kyoto, Japan (Dr Takahashi); Department of Radiology, School of Medicine, Fukushima Medical University, Fukushima, Japan (Dr Ito).,Correspondence: Tetsuya Suhara, MD, PhD, Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, Chiba 263–8555, Japan ()
| |
Collapse
|
13
|
Stößel A, Brox R, Purkayastha N, Hübner H, Hocke C, Prante O, Gmeiner P. Development of molecular tools based on the dopamine D 3 receptor ligand FAUC 329 showing inhibiting effects on drug and food maintained behavior. Bioorg Med Chem 2017; 25:3491-3499. [PMID: 28495386 DOI: 10.1016/j.bmc.2017.04.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/24/2017] [Accepted: 04/27/2017] [Indexed: 12/18/2022]
Abstract
Dopamine D3 receptor-mediated networks have been associated with a wide range of neuropsychiatric diseases, drug addiction and food maintained behavior, which makes D3 a highly promising biological target. The previously described dopamine D3 receptor ligand FAUC 329 (1) showed protective effects against dopamine depletion in a MPTP mouse model of Parkinson's disease. We used the radioligand [18F]2, a [18F]fluoroethoxy substituted analog of the lead compound 1 as a molecular tool for visualization of D3-rich brain regions including the islands of Calleja. Furthermore, structural modifications are reported leading to the pyrimidylpiperazine derivatives 3 and 9 displaying superior subtype selectivity and preference over serotonergic receptors. Evaluation of the lead compound 1 on cocaine-seeking behavior in non-human primates showed a substantial reduction in cocaine self-administration behavior and food intake.
Collapse
Affiliation(s)
- Anne Stößel
- Department of Medicinal Chemistry, Emil Fischer Center, Friedrich-Alexander University, Schuhstraβe 19, D-91052 Erlangen, Germany
| | - Regine Brox
- Department of Medicinal Chemistry, Emil Fischer Center, Friedrich-Alexander University, Schuhstraβe 19, D-91052 Erlangen, Germany
| | - Nirupam Purkayastha
- Department of Medicinal Chemistry, Emil Fischer Center, Friedrich-Alexander University, Schuhstraβe 19, D-91052 Erlangen, Germany
| | - Harald Hübner
- Department of Medicinal Chemistry, Emil Fischer Center, Friedrich-Alexander University, Schuhstraβe 19, D-91052 Erlangen, Germany
| | - Carsten Hocke
- Department of Nuclear Medicine, Ulmenweg 18, D-91054 Erlangen, Germany
| | - Olaf Prante
- Department of Nuclear Medicine, Ulmenweg 18, D-91054 Erlangen, Germany
| | - Peter Gmeiner
- Department of Medicinal Chemistry, Emil Fischer Center, Friedrich-Alexander University, Schuhstraβe 19, D-91052 Erlangen, Germany.
| |
Collapse
|
14
|
Weiland BJ, Zucker RA, Zubieta JK, Heitzeg MM. Striatal dopaminergic reward response relates to age of first drunkenness and feedback response in at-risk youth. Addict Biol 2017; 22:502-512. [PMID: 26732626 DOI: 10.1111/adb.12341] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 10/01/2015] [Accepted: 10/31/2015] [Indexed: 11/30/2022]
Abstract
Dopamine receptor concentrations, primarily in the striatum, are hypothesized to contribute to a developmental imbalance between subcortical and prefrontal control systems in emerging adulthood potentially biasing motivation and increasing risky behaviors. Positron emission tomography studies have found significant reductions in striatal dopamine D2 receptors, and blunted amphetamine-induced dopamine release, in substance users compared with healthy controls. Extant literature is limited and inconsistent concerning vulnerability associated with having a family history of substance abuse (FH+). Some studies have reported familial liability associated with higher dopamine receptor levels, reduced dopamine response to stimulant challenges and decreased response to oral alcohol. However, other reports have failed to find group differences based on family history. We explored the interaction of familial liability and behavioral risk with multi-modal molecular and neural imaging of the dopaminergic system. Forty-four young adult male subjects performed monetary incentive delay tasks during both [11 C]raclopride positron emission tomography and functional magnetic resonance imaging scans. FH+ subjects were identified as low (n = 24) or high risk (n = 9) based on early initiation of drunkenness. FH+ high-risk subjects exhibited heightened striatal dopamine response to monetary reward but did not differ in neural activations compared with FH+ low risk subjects and controls with no familial loading (n = 11). Across all subjects, a negative relationship was found between dopamine release and age of first drunkenness and a positive relationship with neural response to reward receipt. These results suggest that in at-risk individuals, higher dopamine transmission associated with monetary reward may represent a particularly useful neurobiological phenotype.
Collapse
Affiliation(s)
- Barbara J. Weiland
- University of Colorado; Boulder CO, USA
- University of Michigan; Ann Arbor MI, USA
| | | | | | | |
Collapse
|
15
|
Mukherjee J, Majji D, Kaur J, Constantinescu CC, Narayanan TK, Shi B, Nour MT, Pan ML. PET radiotracer development for imaging high-affinity state of dopamine D2 and D3 receptors: Binding studies of fluorine-18 labeled aminotetralins in rodents. Synapse 2016; 71. [PMID: 27864853 DOI: 10.1002/syn.21950] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/15/2016] [Accepted: 11/16/2016] [Indexed: 12/19/2022]
Abstract
Imaging the high-affinity, functional state (HA) of dopamine D2 and D3 receptors has been pursued in PET imaging studies of various brain functions. We report further evaluation of 18 F-5-OH-FPPAT, and the newer 18 F-5-OH-FHXPAT and 18 F-7-OH-FHXPAT. Syntheses of 18 F-5-OH-FHXPAT and 18 F-7-OH-FHXPAT were improved by modifications of our previously reported procedures. Brain slices and brain homogenates from male Sprague-Dawley rats were used with the 3 radiotracers (74-111 kBq/cc). Competition with dopamine (1-100 nM) and Gpp(NH)p (10-50 µM) were carried out to demonstrate binding to dopamine D2 and D3 HA-states and binding kinetics of 18 F-5-OH-FPPAT measured. Ex vivo brain slice autoradiography was carried out on rats administered with 18 F-5-OH-FHXPAT to ascertain HA-state binding. PET/CT imaging in rats and wild type (WT) and D2 knock-out mice were carried out using 18 F-7-OH-FHXPAT (2-37 MBq). Striatum was clearly visualized by the three radiotracers in brain slices and dopamine displaced more than 80% of binding, with dissociation rate in homogenates of 2.2 × 10-2 min-1 for 18 F-5-OH-FPPAT. Treatment with Gpp(NH)p significantly reduced 50-80% striatal binding with faster dissociation rates (5.0 × 10-2 min-1 ), suggesting HA-state binding of 18 F-5-OH-FPPAT and 18 F-5-OH-FHXPAT. Striatal binding of 18 F-5-OH-FHXPAT in ex vivo brain slices were sensitive to Gpp(NH)p, suggesting HA-state binding in vivo. PET binding ratios of 18 F-7-OH-FHXPAT in rat brain were ventral striatum/cerebellum = 2.09 and dorsal striatum/cerebellum = 1.65; similar binding ratios were found in the D2 WT mice. These results suggest that in vivo PET measures of agonists in the brain at least in part reflect binding to the membrane-bound HA-state of the dopamine receptor.
Collapse
Affiliation(s)
- Jogeshwar Mukherjee
- Department of Radiological Sciences, Preclinical Imaging Center, University of California-Irvine, Irvine, California, 92697, USA
| | - Divya Majji
- Department of Radiological Sciences, Preclinical Imaging Center, University of California-Irvine, Irvine, California, 92697, USA
| | - Jasmeet Kaur
- Department of Radiological Sciences, Preclinical Imaging Center, University of California-Irvine, Irvine, California, 92697, USA
| | - Cristian C Constantinescu
- Department of Radiological Sciences, Preclinical Imaging Center, University of California-Irvine, Irvine, California, 92697, USA
| | - Tanjore K Narayanan
- Department of Nuclear Medicine, Kettering Medical Center, Dayton, Ohio, 45429, USA
| | - Bingzhi Shi
- Department of Nuclear Medicine, Kettering Medical Center, Dayton, Ohio, 45429, USA
| | - Mohamed T Nour
- Department of Radiological Sciences, Preclinical Imaging Center, University of California-Irvine, Irvine, California, 92697, USA
| | - Min-Liang Pan
- Department of Radiological Sciences, Preclinical Imaging Center, University of California-Irvine, Irvine, California, 92697, USA
| |
Collapse
|
16
|
Association between DRD2 and DRD3 gene polymorphisms and gastrointestinal symptoms induced by levodopa therapy in Parkinson's disease. THE PHARMACOGENOMICS JOURNAL 2016; 18:196-200. [PMID: 27779245 DOI: 10.1038/tpj.2016.79] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/01/2016] [Accepted: 09/14/2016] [Indexed: 01/14/2023]
Abstract
Levodopa is the most used drug to treat motor symptoms in Parkinson's disease (PD). However, dopaminergic side effects such as nausea and vomiting may occur. Several evidences indicate a major role for dopamine receptors D2 (DRD2) and D3 (DRD3) in emetic activity. The aim of this study was to investigate the relationship of DRD2 rs1799732 and DRD3 rs6280 gene polymorphisms with gastrointestinal (GI) symptoms induced by levodopa in PD patients. Two hundred and seventeen PD patients on levodopa therapy were investigated. DRD2 rs1799732 and DRD3 rs6280 polymorphisms were genotyped by PCR-based methods. Multiple Poisson regression method with robust variance estimators was performed to assess the association between polymorphisms and gastrointestinal symptoms. The analyses showed that DRD2 Ins/Ins (prevalence ratio (PR)=2.374, 95% confidence interval (CI): 1.105-5.100; P=0.027) and DRD3 Ser/Ser genotypes (PR=1.677, 95% CI 1.077-2.611; P=0.022) were independent and predictors of gastrointestinal symptoms associated with levodopa therapy. Despite all the efforts to alleviate GI symptoms, this adverse effect still occurs in PD patients. Pharmacogenetic studies of GI symptoms induced by levodopa therapy have the potential to display new ways to better understand the molecular mechanisms involved in these side effects.
Collapse
|
17
|
Zanatta G, Della Flora Nunes G, Bezerra EM, da Costa RF, Martins A, Caetano EWS, Freire VN, Gottfried C. Two Binding Geometries for Risperidone in Dopamine D3 Receptors: Insights on the Fast-Off Mechanism through Docking, Quantum Biochemistry, and Molecular Dynamics Simulations. ACS Chem Neurosci 2016; 7:1331-1347. [PMID: 27434874 DOI: 10.1021/acschemneuro.6b00074] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Risperidone is an atypical antipsychotic used in the treatment of schizophrenia and of symptoms of irritability associated with autism spectrum disorder (ASD). Its main action mechanism is the blockade of D2-like receptors acting over positive and negative symptoms of schizophrenia with small risk of extrapyramidal symptoms (EPS) at doses corresponding to low/moderate D2 occupancy. Such a decrease in the side effect incidence can be associated with its fast unbinding from D2 receptors in the nigrostriatal region allowing the recovery of dopamine signaling pathways. We performed docking essays using risperidone and the D3 receptor crystallographic data and results suggested two possible distinct orientations for risperidone at the binding pocket. Orientation 1 is more close to the opening of the binding site and has the 6-fluoro-1,2 benzoxazole fragment toward the bottom of the D3 receptor cleft, while orientation 2 is deeper inside the binding pocket with the same fragment toward to the receptor surface. In order to unveil the implications of these two binding orientations, classical molecular dynamics and quantum biochemistry computations within the density functional theory formalism and the molecular fractionation with conjugate caps framework were performed. Quantum mechanics/molecular mechanics suggests that orientation 2 (considering the contribution of Glu90) is slightly more energetically stable than orientation 1 with the main contribution coming from residue Asp110. The residue Glu90, positioned at the opening of the binding site, is closer to orientation 1 than 2, suggesting that it may have a key role in stability through attractive interaction with risperidone. Therefore, although orientations 1 and 2 are both likely to occur, we suggest that the occurrence of the first may contribute to the reduction of side effects in patients taking risperidone due to the reduction of dopamine receptor occupancy in the nigrostriatal region through a mechanism of fast dissociation. The atypical effect may be obtained simply by either delaying D3R full blockage by spatial hindrance of orientation 1 at the binding site or through an effective blockade followed by orientation 1 fast dissociation. While the molecular interpretation suggested in this work shed some light on the potential molecular mechanisms accounting for the reduced extrapyramidal symptoms observed during risperidone treatment, further studies are necessary in order to evaluate the implications of both orientations during the receptor activation/inhibition. Altogether these data highlight important hot spots in the dopamine receptor binding site bringing relevant information for the development of novel/derivative agents with atypical profile.
Collapse
Affiliation(s)
- Geancarlo Zanatta
- Department of Biochemistry, Federal University of Rio Grande do Sul, 90035-003 Porto
Alegre, RS Brazil
| | - Gustavo Della Flora Nunes
- Department of Biochemistry, Federal University of Rio Grande do Sul, 90035-003 Porto
Alegre, RS Brazil
| | - Eveline M. Bezerra
- Post-graduate Program in Pharmaceutical Sciences, Pharmacy Faculty, Federal University of Ceará, 60430-372 Fortaleza, CE Brazil
| | - Roner F. da Costa
- Department of Physics, Universidade Federal Rural do Semi-Árido, 59780-000 Caraúbas, RN Brazil
| | - Alice Martins
- Post-graduate Program in Pharmaceutical Sciences, Pharmacy Faculty, Federal University of Ceará, 60430-372 Fortaleza, CE Brazil
| | - Ewerton W. S. Caetano
- Federal Institute of Education, Science and Technology, 60040-531 Fortaleza, CE Brazil
| | - Valder N. Freire
- Department of Physics, Federal University of Ceará, 60455-760 Fortaleza, CE Brazil
| | - Carmem Gottfried
- Department of Biochemistry, Federal University of Rio Grande do Sul, 90035-003 Porto
Alegre, RS Brazil
| |
Collapse
|
18
|
Jørgensen KN, Nesvåg R, Gunleiksrud S, Raballo A, Jönsson EG, Agartz I. First- and second-generation antipsychotic drug treatment and subcortical brain morphology in schizophrenia. Eur Arch Psychiatry Clin Neurosci 2016; 266:451-60. [PMID: 26547434 DOI: 10.1007/s00406-015-0650-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 10/26/2015] [Indexed: 01/22/2023]
Abstract
Antipsychotic medication may influence brain structure, but to what extent effects of first-generation antipsychotics (FGAs) and second-generation antipsychotics (SGAs) differ is still not clear. Here we aimed to disentangle the effects of FGA and SGA on variation in volumes of subcortical structures in patients with long-term treated schizophrenia. Magnetic resonance images were obtained from 95 patients with schizophrenia and 106 healthy control subjects. Among the patients, 40 received only FGA and 42 received only SGA. FreeSurfer 5.3.0 was used to obtain volumes of 27 subcortical structures as well as total brain volume and estimated intracranial volume. Findings of reduced total brain volume, enlarged ventricular volume and reduced hippocampal volume bilaterally among patients were replicated, largely independent of medication class. In the basal ganglia, FGA users had larger putamen bilaterally and right caudate volume compared to healthy controls, and the right putamen was significantly larger than among SGA users. FGA and SGA users had similar and larger globus pallidus volumes compared to healthy controls. Post hoc analyses revealed that the difference between FGA and SGA could be attributed to smaller volumes in the clozapine users specifically. We therefore conclude that basal ganglia volume enlargements are not specific to FGA.
Collapse
Affiliation(s)
- Kjetil N Jørgensen
- Department of Psychiatric Research, Diakonhjemmet Hospital, P.O. Box 85, 0319, Vinderen, Oslo, Norway. .,NORMENT and K.G. Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Ragnar Nesvåg
- Department of Psychiatric Research, Diakonhjemmet Hospital, P.O. Box 85, 0319, Vinderen, Oslo, Norway.,Department of Genetics, Environment and Mental Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Sindre Gunleiksrud
- Department of Psychiatric Research, Diakonhjemmet Hospital, P.O. Box 85, 0319, Vinderen, Oslo, Norway
| | - Andrea Raballo
- Department of Psychiatric Research, Diakonhjemmet Hospital, P.O. Box 85, 0319, Vinderen, Oslo, Norway.,NORMENT and K.G. Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Erik G Jönsson
- NORMENT and K.G. Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Clinical Neuroscience, HUBIN Project, Karolinska Institutet and Hospital, Stockholm, Sweden
| | - Ingrid Agartz
- Department of Psychiatric Research, Diakonhjemmet Hospital, P.O. Box 85, 0319, Vinderen, Oslo, Norway.,NORMENT and K.G. Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Clinical Neuroscience, HUBIN Project, Karolinska Institutet and Hospital, Stockholm, Sweden
| |
Collapse
|
19
|
Sander CY, Hooker JM, Catana C, Rosen BR, Mandeville JB. Imaging Agonist-Induced D2/D3 Receptor Desensitization and Internalization In Vivo with PET/fMRI. Neuropsychopharmacology 2016; 41:1427-36. [PMID: 26388148 PMCID: PMC4793127 DOI: 10.1038/npp.2015.296] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 09/14/2015] [Accepted: 09/14/2015] [Indexed: 12/23/2022]
Abstract
This study investigated the dynamics of dopamine receptor desensitization and internalization, thereby proposing a new technique for non-invasive, in vivo measurements of receptor adaptations. The D2/D3 agonist quinpirole, which induces receptor internalization in vitro, was administered at graded doses in non-human primates while imaging with simultaneous positron emission tomography (PET) and functional magnetic resonance imaging (fMRI). A pronounced temporal divergence between receptor occupancy and fMRI signal was observed: occupancy remained elevated while fMRI responded transiently. Analogous experiments with an antagonist (prochlorperazine) and a lower-affinity agonist (ropinirole) exhibited reduced temporal dissociation between occupancy and function, consistent with a mechanism of desensitization and internalization that depends upon drug efficacy and affinity. We postulated a model that incorporates internalization into a neurovascular-coupling relationship. This model yielded in vivo desensitization/internalization rates (0.2/min for quinpirole) consistent with published in vitro measurements. Overall, these results suggest that simultaneous PET/fMRI enables characterization of dynamic neuroreceptor adaptations in vivo, and may offer a first non-invasive method for assessing receptor desensitization and internalization.
Collapse
Affiliation(s)
- Christin Y Sander
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA,Harvard Medical School, Boston, MA, USA,A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, 149 Thirteenth Street, Room 2301, Charlestown, MA 02129, USA, Tel: +617 724 1839, Fax: +617 726 7422, E-mail:
| | - Jacob M Hooker
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Ciprian Catana
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Bruce R Rosen
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA,Harvard Medical School, Boston, MA, USA,Health Sciences and Technology, Harvard-MIT, Cambridge, MA, USA
| | - Joseph B Mandeville
- A. A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
20
|
Payer DE, Guttman M, Kish SJ, Tong J, Adams JR, Rusjan P, Houle S, Furukawa Y, Wilson AA, Boileau I. D3 dopamine receptor-preferring [11C]PHNO PET imaging in Parkinson patients with dyskinesia. Neurology 2015; 86:224-30. [PMID: 26718579 DOI: 10.1212/wnl.0000000000002285] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/10/2015] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVE To investigate whether levodopa-induced dyskinesias (LID) are associated with D3 overexpression in levodopa-treated humans with Parkinson disease (PD). METHODS In this case-control study, we used PET with the D3-preferring radioligand [(11)C]-(+)-PHNO to estimate D2/3 receptor binding in patients with levodopa-treated PD with LID (n = 12) and without LID (n = 12), and healthy control subjects matched for age, sex, education, and mental status (n = 18). RESULTS Compared to nondyskinetic patients, those with LID showed heightened [(11)C]-(+)-PHNO binding in the D3-rich globus pallidus. Both PD groups also showed higher binding than controls in the sensorimotor division of the striatum. In contrast, D2/3 binding in the ventral striatum was lower in patients with LID than without, possibly reflecting higher dopamine levels. CONCLUSIONS Dopaminergic abnormalities contributing to LID may include elevated D2/3 binding in globus pallidus, perhaps reflecting D3 receptor upregulation. The findings support therapeutic strategies that target and diminish activity at D3 to prevent LID.
Collapse
Affiliation(s)
- Doris E Payer
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada
| | - Mark Guttman
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada
| | - Stephen J Kish
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada
| | - Junchao Tong
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada
| | - John R Adams
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada
| | - Pablo Rusjan
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada
| | - Sylvain Houle
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada
| | - Yoshiaki Furukawa
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada
| | - Alan A Wilson
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada
| | - Isabelle Boileau
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada.
| |
Collapse
|
21
|
Mukherjee J, Constantinescu CC, Hoang AT, Jerjian T, Majji D, Pan ML. Dopamine D3 receptor binding of (18)F-fallypride: Evaluation using in vitro and in vivo PET imaging studies. Synapse 2015; 69:577-91. [PMID: 26422464 DOI: 10.1002/syn.21867] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/25/2015] [Accepted: 09/19/2015] [Indexed: 12/17/2022]
Abstract
Identification of dopamine D3 receptors (D3R) in vivo is important to understand several brain functions related to addiction. The goal of this work was to identify D3R binding of the dopamine D2 receptor (D2R)/D3R imaging agent, (18)F-fallypride. Brain slices from male Sprague-Dawley rats (n = 6) and New Zealand White rabbits (n = 6) were incubated with (18)F-fallypride and D3R selective agonist (R)-7-OH-DPAT (98-fold D3R selective). Rat slices were also treated with BP 897 (68-fold D3R selective partial agonist) and NGB 2904 (56-fold D3R selective antagonist). In vivo rat studies (n = 6) were done on Inveon PET using 18-37 MBq (18)F-fallypride and drug-induced displacement by (R)-7-OH-DPAT, BP 897 and NGB 2904. PET/CT imaging of wild type (WT, n = 2) and D2R knock-out (KO, n = 2) mice were carried out with (18)F-fallypride. (R)-7-OH-DPAT displaced binding of (18)F-fallypride, both in vitro and in vivo. In vitro, at 10 nM (R)-7-OH-DPAT, (18)F-fallypride binding in the rat ventral striatum (VST) and dorsal striatum (DST) and rabbit nucleus accumbens were reduced by ∼10-15%. At 10 μM (R)-7-OH-DPAT all regions in rat and rabbit were reduced by ≥85%. In vivo reductions for DST and VST before and after (R)-7-OH-DPAT were: low-dose (0.015 mg kg(-1)) DST -22%, VST -29%; high-dose (1.88 mg kg(-1)) DST -58%, VST -77%, suggesting D3R/D2R displacement. BP 897 and NGB 2904 competed with (18)F-fallypride in vitro, but unlike BP 897, NGB 2904 did not displace (18)F-fallypride in vivo. The D2R KO mice lacked (18)F-fallypride binding in the DST. In summary, our findings suggest that up to 20% of (18)F-fallypride may be bound to D3R sites in vivo.
Collapse
Affiliation(s)
- Jogeshwar Mukherjee
- Department of Radiological Sciences, Preclinical Imaging, University of California, Irvine, California, 92697
| | - Cristian C Constantinescu
- Department of Radiological Sciences, Preclinical Imaging, University of California, Irvine, California, 92697
| | - Angela T Hoang
- Department of Radiological Sciences, Preclinical Imaging, University of California, Irvine, California, 92697
| | - Taleen Jerjian
- Department of Radiological Sciences, Preclinical Imaging, University of California, Irvine, California, 92697
| | - Divya Majji
- Department of Radiological Sciences, Preclinical Imaging, University of California, Irvine, California, 92697
| | - Min-Liang Pan
- Department of Radiological Sciences, Preclinical Imaging, University of California, Irvine, California, 92697
| |
Collapse
|
22
|
Chronic Back Pain Is Associated with Alterations in Dopamine Neurotransmission in the Ventral Striatum. J Neurosci 2015; 35:9957-65. [PMID: 26156996 DOI: 10.1523/jneurosci.4605-14.2015] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Back pain is common in the general population, but only a subgroup of back pain patients develops a disabling chronic pain state. The reasons for this are incompletely understood, but recent evidence implies that both preexisting and pain-related variations in the structure and function of the nervous system may contribute significantly to the development of chronic pain. Here, we addressed the role of striatal dopamine (DA) D2/D3 receptor (D2/D3R) function in chronic non-neuropathic back pain (CNBP) by comparing CNBP patients and healthy controls using PET and the D2/D3R-selective radioligand [(11)C]raclopride. D2/D3R availability was measured at baseline and during a pain challenge, yielding in vivo measures of receptor availability (binding potential, BPND) and DA release (change in BPND from baseline to activated state). At baseline, CNBP patients demonstrated reductions in D2/D3R BPND in the ventral striatum compared with controls. These reductions were associated with greater positive affect scores and pain tolerance measures. The reductions in D2/D3R BPND were also correlated with μ-opioid receptor BPND and pain-induced endogenous opioid system activation in the amygdala, further associated with measures of positive affect, the affective component of back pain and pain tolerance. During the pain challenge, lower magnitudes of DA release, and therefore D2/D3R activation, were also found in the ventral striatum in the CNBP sample compared with controls. Our results show that CNBP is associated with adaptations in ventral striatal D2/D3R function, which, together with endogenous opioid system function, contribute to the sensory and affective-motivational features of CNBP. SIGNIFICANCE STATEMENT The neural systems that underlie chronic pain remain poorly understood. Here, using PET, we provide insight into the molecular mechanisms that regulate sensory and affective dimensions of pain in chronic back pain patients. We found that patients with back pain have alterations in brain dopamine function that are associated with measures of pain sensitivity and affective state, but also with brain endogenous opioid system functional measures. These findings suggest that brain dopamine-opioid interactions are involved in the pathophysiology of chronic pain, which has potential therapeutic implications. Our results may also help to explain individual variation in susceptibility to opioid medication misuse and eventual addiction in the context of chronic pain.
Collapse
|
23
|
Novel dimensions of D3 receptor function: Focus on heterodimerisation, transactivation and allosteric modulation. Eur Neuropsychopharmacol 2015; 25:1470-9. [PMID: 25453482 DOI: 10.1016/j.euroneuro.2014.09.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 09/24/2014] [Indexed: 02/08/2023]
Abstract
The brain׳s complexity derives not only from the way the intricate network of neurons is wired, but also by protein complexes that recognize and decode chemical information. G protein-coupled receptors (GPCRs) represent the most abundant family of proteins mediating neurotransmission in the brain, and their ability to form homo- and heteromers which amplifies the scope for synaptic communication and fine-tuning. Dopamine receptors are important drug targets and members of both the D1/D5 and D2/D3/D4 receptor families form homo- and heteromers. The present article focuses on D3 receptor homo- and heteromers, in particular, those formed in association with their D2 counterparts. We highlight the binding profiles and mechanisms of interaction with D3-D3 homomers and D3-D2 heteromers of: first, the PET ligand and potent agonist [(11)C]-(+)-PHNO; second, the novel, bitopic/allosteric dopamine D3 receptor antagonist, SB269,652; and third, diverse partial agonists like antipsychotic and aripiprazole. Molecular mechanisms of interplay between the two protomers of heteromeric D3-D2 complexes are likewise discussed: for example, "transactivation", whereby recruitment of one member of a heteromer harnesses signalling pathways is normally coupled to the other protomer. Finally, D1 receptor heteromers are also taken into consideration in deciphering the nature of interfaces required to stabilize dimeric assemblies and permit their interaction with G proteins. Improved understanding of D3 as well as D2 and D1 receptor complexes should yield important insights into their physiological roles and pathological significance, and permit the development of novel drug classes with potentially distinctive functional profiles and improved therapeutic windows.
Collapse
|
24
|
Seeman P. Parkinson's disease treatment may cause impulse-control disorder via dopamine D3 receptors. Synapse 2015; 69:183-9. [DOI: 10.1002/syn.21805] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 12/23/2014] [Accepted: 01/09/2015] [Indexed: 01/15/2023]
Affiliation(s)
- Philip Seeman
- Departments of Pharmacology and Psychiatry; Faculty of Medicine, University of Toronto; 260 Heath Street West, Unit 605, Toronto Ontario M5P 3L6 Canada
| |
Collapse
|
25
|
Zanatta G, Nunes G, Bezerra EM, da Costa RF, Martins A, Caetano EWS, Freire VN, Gottfried C. Antipsychotic haloperidol binding to the human dopamine D3 receptor: beyond docking through QM/MM refinement toward the design of improved schizophrenia medicines. ACS Chem Neurosci 2014; 5:1041-54. [PMID: 25181639 DOI: 10.1021/cn500111e] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
As the dopamine D3R receptor is a promising target for schizophrenia treatment, an improved understanding of the binding of existing antipsychotics to this receptor is crucial for the development of new potent and more selective therapeutic agents. In this work, we have used X-ray cocrystallization data of the antagonist eticlopride bound to D3R as a template to predict, through docking essays, the placement of the typical antipsychotic drug haloperidol at the D3R receptor binding site. Afterward, classical and quantum mechanics/molecular mechanics (QM/MM) computations were employed to improve the quality of the docking calculations, with the QM part of the simulations being accomplished by using the density functional theory (DFT) formalism. After docking, the calculated QM improved total interaction energy EQMDI = -170.1 kcal/mol was larger (in absolute value) than that obtained with classical molecular mechanics improved (ECLDI = -156.3 kcal/mol) and crude docking (ECRDI = -137.6 kcal/mol) procedures. The QM/MM computations reveal the pivotal role of the Asp110 amino acid residue in the D3R haloperidol binding, followed by Tyr365, Phe345, Ile183, Phe346, Tyr373, and Cys114. Besides, it highlights the relevance of the haloperidol hydroxyl group axial orientation, which interacts with the Tyr365 and Thr369 residues, enhancing its binding to dopamine receptors. Finally, our computations indicate that functional substitutions in the 4-clorophenyl and in the 4-hydroxypiperidin-1-yl fragments (such as C3H and C12H hydrogen replacement by OH or COOH) can lead to haloperidol derivatives with distinct dopamine antagonism profiles. The results of our work are a first step using in silico quantum biochemical design as means to impact the discovery of new medicines to treat schizophrenia.
Collapse
Affiliation(s)
- Geancarlo Zanatta
- Department
of Biochemistry, Federal University of Rio Grande do Sul, 90035-003 Porto Alegre, RS Brazil
| | - Gustavo Nunes
- Department
of Biochemistry, Federal University of Rio Grande do Sul, 90035-003 Porto Alegre, RS Brazil
| | - Eveline M. Bezerra
- Post-graduate
Program in Pharmaceutical Sciences, Pharmacy Faculty, Federal University of Ceará, 60430-372 Fortaleza, CE Brazil
| | - Roner F. da Costa
- Department
of Physics, Universidade Federal Rural do Semi-Árido, 59780-000 Caraúbas, RN Brazil
| | - Alice Martins
- Post-graduate
Program in Pharmaceutical Sciences, Pharmacy Faculty, Federal University of Ceará, 60430-372 Fortaleza, CE Brazil
| | - Ewerton W. S. Caetano
- Federal Institute of Education, Science and Technology, 60040-531 Fortaleza, CE Brazil
| | - Valder N. Freire
- Department
of Physics, Federal University of Ceará, 60455-760 Fortaleza, CE Brazil
| | - Carmem Gottfried
- Department
of Biochemistry, Federal University of Rio Grande do Sul, 90035-003 Porto Alegre, RS Brazil
| |
Collapse
|
26
|
Weiland BJ, Heitzeg MM, Zald D, Cummiford C, Love T, Zucker RA, Zubieta JK. Relationship between impulsivity, prefrontal anticipatory activation, and striatal dopamine release during rewarded task performance. Psychiatry Res 2014; 223:244-52. [PMID: 24969539 PMCID: PMC4136473 DOI: 10.1016/j.pscychresns.2014.05.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 05/05/2014] [Accepted: 05/26/2014] [Indexed: 11/29/2022]
Abstract
Impulsivity, and in particular the negative urgency aspect of this trait, is associated with poor inhibitory control when experiencing negative emotion. Individual differences in aspects of impulsivity have been correlated with striatal dopamine D2/D3 receptor availability and function. This multi-modal pilot study used both positron emission tomography (PET) and functional magnetic resonance imaging (fMRI) to evaluate dopaminergic and neural activity, respectively, using modified versions of the monetary incentive delay task. Twelve healthy female subjects underwent both scans and completed the NEO Personality Inventory Revised to assess Impulsiveness (IMP). We examined the relationship between nucleus accumbens (NAcc) dopaminergic incentive/reward release, measured as a change in D2/D3 binding potential between neutral and incentive/reward conditions with [(11)C]raclopride PET, and blood oxygen level-dependent (BOLD) activation elicited during the anticipation of rewards, measured with fMRI. Left NAcc incentive/reward dopaminergic release correlated with anticipatory reward activation within the medial prefrontal cortex (mPFC), left angular gyrus, mammillary bodies, and left superior frontal cortex. Activation in the mPFC negatively correlated with IMP and mediated the relationship between IMP and incentive/reward dopaminergic release in left NAcc. The mPFC, with a regulatory role in learning and valuation, may influence dopamine incentive/reward release.
Collapse
Affiliation(s)
- Barbara J Weiland
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, USA; Department of Psychiatry, The University of Michigan, Ann Arbor, MI, USA.
| | - Mary M Heitzeg
- Department of Psychiatry, The University of Michigan, Ann Arbor, MI, USA
| | - David Zald
- Department of Psychiatry, Vanderbilt University, Nashville, TN, USA
| | - Chelsea Cummiford
- Department of Psychiatry, The University of Michigan, Ann Arbor, MI, USA
| | - Tiffany Love
- Department of Psychiatry, The University of Michigan, Ann Arbor, MI, USA
| | - Robert A Zucker
- Department of Psychiatry, The University of Michigan, Ann Arbor, MI, USA
| | - Jon-Kar Zubieta
- Department of Psychiatry, The University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
27
|
The influence of genetic variants on striatal dopamine transporter and D2 receptor binding after TBI. J Cereb Blood Flow Metab 2014; 34:1328-39. [PMID: 24849661 PMCID: PMC4126093 DOI: 10.1038/jcbfm.2014.87] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 03/12/2014] [Accepted: 04/21/2014] [Indexed: 12/24/2022]
Abstract
Dopamine (DA) neurotransmission influences cognition and recovery after traumatic brain injury (TBI). We explored whether functional genetic variants affecting the DA transporter (DAT) and D2 receptor (DRD2) impacted in vivo dopaminergic binding with positron emission tomography (PET) using [(11)C]βCFT and [(11)C]raclopride. We examined subjects with moderate/severe TBI (N=12) ∼1 year post injury and similarly matched healthy controls (N=13). The variable number of tandem repeat polymorphism within the DAT gene and the TaqI restriction fragment length polymorphism near the DRD2 gene were assessed. TBI subjects had age-adjusted DAT-binding reductions in the caudate, putamen, and ventral striatum, and modestly increased D2 binding in ventral striatum versus controls. Despite small sample sizes, multivariate analysis showed lower caudate and putamen DAT binding among DAT 9-allele carriers and DRD2 A2/A2 homozygotes with TBI versus controls with the same genotype. Among TBI subjects, 9-allele carriers had lower caudate and putamen binding than 10/10 homozygotes. This PET study suggests a hypodopaminergic environment and altered DRD2 autoreceptor DAT interactions that may influence DA transmission after TBI. Future work will relate these findings to cognitive performance; future studies are required to determine how DRD2/DAT1 genotype and DA-ligand binding are associated with neurostimulant response and TBI recovery.
Collapse
|
28
|
van Coevorden-Hameete MH, de Graaff E, Titulaer MJ, Hoogenraad CC, Sillevis Smitt PAE. Molecular and cellular mechanisms underlying anti-neuronal antibody mediated disorders of the central nervous system. Autoimmun Rev 2014; 13:299-312. [PMID: 24225076 DOI: 10.1016/j.autrev.2013.10.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Accepted: 10/30/2013] [Indexed: 12/31/2022]
Abstract
Over the last decade multiple autoantigens located on the plasma membrane of neurons have been identified. Neuronal surface antigens include molecules directly involved in neurotransmission and excitability. Binding of the antibody to the antigen may directly alter the target protein's function, resulting in neurological disorders. The often striking reversibility of symptoms following early aggressive immunotherapy supports a pathogenic role for autoantibodies to neuronal surface antigens. In order to better understand and treat these neurologic disorders it is important to gain insight in the underlying mechanisms of antibody pathogenicity. In this review we discuss the clinical, circumstantial, in vitro and in vivo evidence for neuronal surface antibody pathogenicity and the possible underlying cellular and molecular mechanisms. This review shows that antibodies to neuronal surface antigens are often directed at conformational epitopes located in the extracellular domain of the antigen. The conformation of the epitope can be affected by specific posttranslational modifications. This may explain the distinct clinical phenotypes that are seen in patients with antibodies to antigens that are expressed throughout the brain. Furthermore, it is likely that there is a heterogeneous antibody population, consisting of different IgG subtypes and directed at multiple epitopes located in an immunogenic region. Binding of these antibodies may result in different pathophysiological mechanisms occurring in the same patient, together contributing to the clinical syndrome. Unraveling the predominant mechanism in each distinct antigen could provide clues for therapeutic interventions.
Collapse
Affiliation(s)
- M H van Coevorden-Hameete
- Department of Biology, Division of Cell Biology, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| | - E de Graaff
- Department of Biology, Division of Cell Biology, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| | - M J Titulaer
- Department of Neurology, Erasmus MC, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| | - C C Hoogenraad
- Department of Biology, Division of Cell Biology, Utrecht University, Padualaan 8, 3584 CH, Utrecht, The Netherlands.
| | - P A E Sillevis Smitt
- Department of Neurology, Erasmus MC, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands.
| |
Collapse
|
29
|
Kumar R, Farahvar S, Ogren JA, Macey PM, Thompson PM, Woo MA, Yan-Go FL, Harper RM. Brain putamen volume changes in newly-diagnosed patients with obstructive sleep apnea. NEUROIMAGE-CLINICAL 2014; 4:383-91. [PMID: 24567910 PMCID: PMC3930100 DOI: 10.1016/j.nicl.2014.01.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 01/17/2014] [Accepted: 01/21/2014] [Indexed: 11/24/2022]
Abstract
Obstructive sleep apnea (OSA) is accompanied by cognitive, motor, autonomic, learning, and affective abnormalities. The putamen serves several of these functions, especially motor and autonomic behaviors, but whether global and specific sub-regions of that structure are damaged is unclear. We assessed global and regional putamen volumes in 43 recently-diagnosed, treatment-naïve OSA (age, 46.4 ± 8.8 years; 31 male) and 61 control subjects (47.6 ± 8.8 years; 39 male) using high-resolution T1-weighted images collected with a 3.0-Tesla MRI scanner. Global putamen volumes were calculated, and group differences evaluated with independent samples t-tests, as well as with analysis of covariance (covariates; age, gender, and total intracranial volume). Regional differences between groups were visualized with 3D surface morphometry-based group ratio maps. OSA subjects showed significantly higher global putamen volumes, relative to controls. Regional analyses showed putamen areas with increased and decreased tissue volumes in OSA relative to control subjects, including increases in caudal, mid-dorsal, mid-ventral portions, and ventral regions, while areas with decreased volumes appeared in rostral, mid-dorsal, medial-caudal, and mid-ventral sites. Global putamen volumes were significantly higher in the OSA subjects, but local sites showed both higher and lower volumes. The appearance of localized volume alterations points to differential hypoxic or perfusion action on glia and other tissues within the structure, and may reflect a stage in progression of injury in these newly-diagnosed patients toward the overall volume loss found in patients with chronic OSA. The regional changes may underlie some of the specific deficits in motor, autonomic, and neuropsychologic functions in OSA. Global and regional putamen volumes were examined in newly-diagnosed OSA. Global volumes are higher, but subareas showed increases and decreases. The volume increases suggest transient tissue swelling from hypoxic action. Altered sites likely contribute to motor and other functional deficits in OSA.
Collapse
Key Words
- 3D surface morphometry
- 3D, Three dimensional
- AHI, Apnea–hypopnea index
- Autonomic
- BAI, Beck Anxiety Inventory
- BDI-II, Beck Depression Inventory II
- Basal ganglia
- CSF, Cerebrospinal fluid
- Cognition
- ESS, Epworth Sleepiness Scale
- FA, Flip angle
- FOV, Field of view
- GRAPPA, Generalized autocalibrating partially parallel acquisition
- Intermittent hypoxia
- MNI, Montreal Neurological Institute
- MPRAGE, Magnetization prepared rapid acquisition gradient-echo
- MRI, Magnetic resonance imaging
- Magnetic resonance imaging
- Motor
- OSA, Obstructive sleep apnea
- PD, Proton density
- PSQI, Pittsburgh Sleep Quality Index
- TE, Echo time
- TIV, Total intracranial volume
- TR, Repetition time
Collapse
Affiliation(s)
- Rajesh Kumar
- Department of Anesthesiology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, USA ; Department of Radiological Sciences, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, USA ; The Brain Research Institute, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Salar Farahvar
- Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Jennifer A Ogren
- UCLA School of Nursing, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Paul M Macey
- UCLA School of Nursing, University of California at Los Angeles, Los Angeles, CA 90095, USA ; The Brain Research Institute, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Paul M Thompson
- Department of Neurology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, USA ; Department of Psychiatry, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Mary A Woo
- UCLA School of Nursing, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Frisca L Yan-Go
- Department of Neurology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Ronald M Harper
- Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA 90095, USA ; The Brain Research Institute, University of California at Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
30
|
Hocke C, Cumming P, Maschauer S, Kuwert T, Gmeiner P, Prante O. Biodistribution studies of two 18F-labeled pyridinylphenyl amides as subtype selective radioligands for the dopamine D3 receptor. Nucl Med Biol 2013; 41:223-8. [PMID: 24480780 DOI: 10.1016/j.nucmedbio.2013.12.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 12/03/2013] [Accepted: 12/12/2013] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Dopamine D3 receptors are implicated in various neuropsychiatric diseases, drug abuse and alcoholism, but specific agents for D3 molecular imaging are lacking. We evaluated two in vitro selective fluorine-18-labeled radioligand candidates ([(18)F]5 and [(18)F]6) for positron emission tomography (PET) imaging of D3 receptor availability in the brain. METHODS Biodistribution was evaluated in Sprague-Dawley rats using ex vivo autoradiography and small-animal PET. Protein binding studies were conducted in human plasma and cerebrospinal fluid. RESULTS [(18)F]5 showed rapid blood-brain barrier penetration and fast washout after intravenous injection, whereas the rat brain penetration of [(18)F]6 was lower. The total distribution volume (VT) of [(18)F]5 was 20-26 mL g(-1) throughout brain. Co-injection with the D3 antagonist BP897 resulted in globally increased cerebral washout of [(18)F]5 and [(18)F]6, but SUV analysis and parametric mapping of binding potential (BPND) relative to the cerebellum did not reveal specific binding of either ligand in D3-rich brain regions, i.e. the ventral striatum. However, there was substantial displaceable binding of [(18)F]5, and to a lesser extent [(18)F]6, in the pituitary. CONCLUSION These radioligands reveal dopamine D3 receptors in the pituitary, but are not suitable for PET imaging of in brain, possibly due to low specific signal relative to the globally high VT.
Collapse
Affiliation(s)
- Carsten Hocke
- Molecular Imaging and Radiochemistry, Nuclear Medicine Clinic, Friedrich-Alexander University, Schwabachanlage 6, D-91054 Erlangen, Germany.
| | - Paul Cumming
- Molecular Imaging and Radiochemistry, Nuclear Medicine Clinic, Friedrich-Alexander University, Schwabachanlage 6, D-91054 Erlangen, Germany
| | - Simone Maschauer
- Molecular Imaging and Radiochemistry, Nuclear Medicine Clinic, Friedrich-Alexander University, Schwabachanlage 6, D-91054 Erlangen, Germany
| | - Torsten Kuwert
- Molecular Imaging and Radiochemistry, Nuclear Medicine Clinic, Friedrich-Alexander University, Schwabachanlage 6, D-91054 Erlangen, Germany
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Emil Fischer Center, Friedrich-Alexander University, Schuhstrasse 19, D-91052 Erlangen, Germany
| | - Olaf Prante
- Molecular Imaging and Radiochemistry, Nuclear Medicine Clinic, Friedrich-Alexander University, Schwabachanlage 6, D-91054 Erlangen, Germany
| |
Collapse
|
31
|
Gross G, Drescher K. The role of dopamine D(3) receptors in antipsychotic activity and cognitive functions. Handb Exp Pharmacol 2013:167-210. [PMID: 23027416 DOI: 10.1007/978-3-642-25758-2_7] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Dopamine D(3) receptors have a pre- and postsynaptic localization in brain stem nuclei, limbic parts of the striatum, and cortex. Their widespread influence on dopamine release, on dopaminergic function, and on several other neurotransmitters makes them attractive targets for therapeutic intervention. The signaling pathways of D(3) receptors are distinct from those of other members of the D(2)-like receptor family. There is increasing evidence that D(3) receptors can form heteromers with dopamine D(1), D(2), and probably other G-protein-coupled receptors. The functional consequences remain to be characterized in more detail but might open new interesting pharmacological insight and opportunities. In terms of behavioral function, D(3) receptors are involved in cognitive, social, and motor functions, as well as in filtering and sensitization processes. Although the role of D(3) receptor blockade for alleviating positive symptoms is still unsettled, selective D(3) receptor antagonism has therapeutic features for schizophrenia and beyond as demonstrated by several animal models: improved cognitive function, emotional processing, executive function, flexibility, and social behavior. D(3) receptor antagonism seems to contribute to atypicality of clinically used antipsychotics by reducing extrapyramidal motor symptoms; has no direct influence on prolactin release; and does not cause anhedonia, weight gain, or metabolic dysfunctions. Unfortunately, clinical data with new, selective D(3) antagonists are still incomplete; their cognitive effects have only been communicated in part. In vitro, virtually all clinically used antipsychotics are not D(2)-selective but also have affinity for D(3) receptors. The exact D(3) receptor occupancies achieved in patients, particularly in cortical areas, are largely unknown, mainly because only nonselective or agonist PET tracers are currently available. It is unlikely that a degree of D(3) receptor antagonism optimal for antipsychotic and cognitive function can be achieved with existing antipsychotics. Therefore, selective D(3) antagonism represents a promising mechanism still to be fully exploited for the treatment of schizophrenia, cognitive deficits in schizophrenia, and comorbid conditions such as substance abuse.
Collapse
Affiliation(s)
- Gerhard Gross
- Abbott, Neuroscience Research, Ludwigshafen, Germany.
| | | |
Collapse
|
32
|
Abstract
Neural systems that identify and respond to salient stimuli are critical for survival in a complex and changing environment. In addition, interindividual differences, including genetic variation and hormonal and metabolic status likely influence the behavioral strategies and neuronal responses to environmental challenges. Here, we examined the relationship between leptin allelic variation and plasma leptin levels with DAD2/3R availability in vivo as measured with [(11)C]raclopride PET at baseline and during a standardized pain stress challenge. Allelic variation in the leptin gene was associated with varying levels of dopamine release in response to the pain stressor, but not with baseline D2/3 receptor availability. Circulating leptin was also positively associated with stress-induced dopamine release. These results show that leptin serves as a regulator of neuronal function in humans and provides an etiological mechanism for differences in dopamine neurotransmission in response to salient stimuli as related to metabolic function. The capacity for leptin to influence stress-induced dopaminergic function is of importance for pathological states where dopamine is thought to play an integral role, such as mood, substance-use disorders, eating disorders, and obesity.
Collapse
|
33
|
Kodaka F, Ito H, Kimura Y, Fujie S, Takano H, Fujiwara H, Sasaki T, Nakayama K, Halldin C, Farde L, Suhara T. Test-retest reproducibility of dopamine D2/3 receptor binding in human brain measured by PET with [11C]MNPA and [11C]raclopride. Eur J Nucl Med Mol Imaging 2012; 40:574-9. [PMID: 23238524 DOI: 10.1007/s00259-012-2312-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 11/21/2012] [Indexed: 01/28/2023]
Abstract
PURPOSE Dopamine D receptors (DRs) have two affinity states for endogenous dopamine, referred to as high-affinity state (D ), which has a high affinity for endogenous dopamine, and low-affinity state (D ). The density of D can be measured with (R)-2-CHO-N-n-propylnorapomorphine ([C]MNPA), while total density of D and D (DRs) can be measured with [C]raclopride using positron emission tomography (PET). Thus, the ratio of the binding potential (BP) of [C]MNPA to that of [C]raclopride ([C]MNPA/[C]raclopride) may reflect the proportion of the density of D to that of DRs. In the caudate and putamen, [C]MNPA/[C]raclopride reflects the proportion of the density of D to that of DRs. To evaluate the reliability of the PET paradigm with [C]MNPA and [C]raclopride, we investigated the test-retest reproducibility of non-displaceable BP (BP ) measured with [C]MNPA and of [C]MNPA/[C]raclopride in healthy humans. METHODS Eleven healthy male volunteers underwent two sets of PET studies on separate days that each included [C]MNPA and [C]raclopride scans. BP values in the caudate and putamen were calculated. Test-retest reproducibility of BP of [C]MNPA and [C]MNPA/[C]raclopride was assessed by intra-subject variability (absolute variability) and test-retest reliability (intraclass correlation coefficient: ICC). RESULTS The absolute variability of [C]MNPA BP was 5.30 ± 3.96 % and 12.3 ± 7.95 % and the ICC values of [C]MNPA BP were 0.72 and 0.82 in the caudate and putamen, respectively. The absolute variability of [C]MNPA/[C]raclopride was 6.11 ± 3.68 % and 11.60 ± 5.70 % and the ICC values of [C]MNPA/[C]raclopride were 0.79 and 0.80 in the caudate and putamen, respectively. CONCLUSION In the present preliminary study, the test-retest reproducibility of BP of [C]MNPA and of [C]MNPA/[C]raclopride was reliable in the caudate and putamen.
Collapse
Affiliation(s)
- Fumitoshi Kodaka
- Molecular Neuroimaging Program, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Searle GE, Beaver JD, Tziortzi A, Comley RA, Bani M, Ghibellini G, Merlo-Pich E, Rabiner EA, Laruelle M, Gunn RN. Mathematical modelling of [¹¹C]-(+)-PHNO human competition studies. Neuroimage 2012. [PMID: 23207573 DOI: 10.1016/j.neuroimage.2012.11.033] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
The D(2)/D(3) agonist radioligand [(11)C]-(+)-PHNO is currently the most suitable D(3) imaging agent available, despite its limited selectivity for the D(3) over the D(2). Given the collocation of D(2) and D(3) receptors, and generally higher densities of D(2), the separation of D(2) and D(3) information from [(11)C]-(+)-PHNO PET data are somewhat complex. This complexity is compounded by recent data suggesting that [(11)C]-(+)-PHNO PET scans might be routinely performed in non-tracer conditions (with respect to D(3) receptors), and that the cerebellum (used as a reference region) might manifest some displaceable binding signal. Here we present the modelling and analysis of data from two human studies which employed an adequate dose range of selective D(3) antagonists (GSK598809 and GSK618334) to interrogate the [(11)C]-(+)-PHNO PET signal. Models describing the changes observed in the PET volume of distribution (V(T)) and binding potential (BP(ND)) were used to identify and quantify a [(11)C]-(+)-PHNO mass dose effect at the D(3), and displaceable signal in the cerebellum, as well as providing refined estimates of regional D(3) fractions of [(11)C]-(+)-PHNO BP(ND). The dose of (+)-PHNO required to occupy half of the available D(3) receptors (ED(50)(PHNO,D3)) was estimated as 40ng/kg, and the cerebellum BP(ND) was estimated as 0.40. These findings confirm that [(11)C]-(+)-PHNO human PET studies are in fact routinely performed under non-tracer conditions. This suggests that (+)-PHNO injection masses should be minimised and tightly controlled in order to mitigate the mass dose effect. The specific binding detected in the cerebellum was modest but could have a significant effect, for example on estimates of D(3) potency in drug occupancy studies. A range of methods for the analysis of future [(11)C]-(+)-PHNO data, incorporating models for the effects quantified here, were developed and evaluated. The comparisons and conclusions drawn from these can inform the design and analysis of future PET studies with [(11)C]-(+)-PHNO.
Collapse
Affiliation(s)
- Graham E Searle
- GlaxoSmithKline Clinical Imaging Centre, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Noam Y, Phan L, McClelland S, Manders EM, Ehrengruber MU, Wadman WJ, Baram TZ, Chen Y. Distinct regional and subcellular localization of the actin-binding protein filamin A in the mature rat brain. J Comp Neurol 2012; 520:3013-34. [PMID: 22434607 DOI: 10.1002/cne.23106] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Filamin A (FLNa) is an actin-binding protein that regulates cell motility, adhesion, and elasticity by cross-linking filamentous actin. Additional roles of FLNa include regulation of protein trafficking and surface expression. Although the functions of FLNa during brain development are well studied, little is known on its expression, distribution, and function in the adult brain. Here we characterize in detail the neuroanatomical distribution and subcellular localization of FLNa in the mature rat brain, by using two antisera directed against epitopes at either the N' or the C' terminus of the protein, further validated by mRNA expression. FLNa was widely and selectively expressed throughout the brain, and the intensity of immunoreactivity was region dependent. The most intensely FLNa-labeled neurons were found in discrete neuronal systems, including basal forebrain structures, anterior nuclear group of thalamus, and hypothalamic parvocellular neurons. Pyramidal neurons in neocortex and hippocampus and magnocellular cells in basolateral amygdaloid nucleus were also intensely FLNa immunoreactive, and strong FLNa labeling was evident in the pontine and medullary raphe nuclei and in sensory and spinal trigeminal nuclei. The subcellular localization of FLNa was evaluated in situ as well as in primary hippocampal neurons. Punctate expression was found in somata and along the dendritic shaft, but FLNa was not detected in dendritic spines. These subcellular distribution patterns were recapitulated in hippocampal and neocortical pyramidal neurons in vivo. The characterization of the expression and subcellular localization of FLNa may provide new clues to the functional roles of this cytoskeletal protein in the adult brain.
Collapse
Affiliation(s)
- Yoav Noam
- Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, California 92697-4475, USA
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Striatal dopamine D₂/D₃ receptors mediate response inhibition and related activity in frontostriatal neural circuitry in humans. J Neurosci 2012; 32:7316-24. [PMID: 22623677 DOI: 10.1523/jneurosci.4284-11.2012] [Citation(s) in RCA: 182] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Impulsive behavior is thought to reflect a traitlike characteristic that can have broad consequences for an individual's success and well-being, but its neurobiological basis remains elusive. Although striatal dopamine D₂-like receptors have been linked with impulsive behavior and behavioral inhibition in rodents, a role for D₂-like receptor function in frontostriatal circuits mediating inhibitory control in humans has not been shown. We investigated this role in a study of healthy research participants who underwent positron emission tomography with the D₂/D₃ dopamine receptor ligand [¹⁸F]fallypride and BOLD fMRI while they performed the Stop-signal Task, a test of response inhibition. Striatal dopamine D₂/D₃ receptor availability was negatively correlated with speed of response inhibition (stop-signal reaction time) and positively correlated with inhibition-related fMRI activation in frontostriatal neural circuitry. Correlations involving D₂/D₃ receptor availability were strongest in the dorsal regions (caudate and putamen) of the striatum, consistent with findings of animal studies relating dopamine receptors and response inhibition. The results suggest that striatal D₂-like receptor function in humans plays a major role in the neural circuitry that mediates behavioral control, an ability that is essential for adaptive responding and is compromised in a variety of common neuropsychiatric disorders.
Collapse
|
37
|
Love TM, Enoch MA, Hodgkinson CA, Pecina M, Mickey B, Koeppe RA, Stohler CS, Goldman D, Zubieta JK. Oxytocin gene polymorphisms influence human dopaminergic function in a sex-dependent manner. Biol Psychiatry 2012; 72:198-206. [PMID: 22418012 PMCID: PMC3392442 DOI: 10.1016/j.biopsych.2012.01.033] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2011] [Revised: 01/13/2012] [Accepted: 01/26/2012] [Indexed: 02/06/2023]
Abstract
BACKGROUND Oxytocin, classically involved in social and reproductive activities, is increasingly recognized as an antinociceptive and anxiolytic agent, effects which may be mediated via oxytocin's interactions with the dopamine system. Thus, genetic variation within the oxytocin gene (OXT) is likely to explain variability in dopamine-related stress responses. As such, we examined how OXT variation is associated with stress-induced dopaminergic neurotransmission in a healthy human sample. METHODS Fifty-five young healthy volunteers were scanned using [¹¹C]raclopride positron emission tomography while they underwent a standardized physical and emotional stressor that consisted of moderate levels of experimental sustained deep muscle pain, and a baseline, control state. Four haplotype tagging single nucleotide polymorphisms located in regions near OXT were genotyped. Measures of pain, affect, anxiety, well-being and interpersonal attachment were also assessed. RESULTS Female rs4813625 C allele carriers demonstrated greater stress-induced dopamine release, measured as reductions in receptor availability from baseline to the pain-stress condition relative to female GG homozygotes. No significant differences were detected among males. We also observed that female rs4813625 C allele carriers exhibited higher attachment anxiety, higher trait anxiety and lower emotional well-being scores. In addition, greater stress-induced dopamine release was associated with lower emotional well-being scores in female rs4813625 C allele carriers. CONCLUSIONS Our results suggest that variability within the oxytocin gene appear to explain interindividual differences in dopaminergic responses to stress, which are shown to be associated with anxiety traits, including those linked to attachment style, as well as emotional well-being in women.
Collapse
Affiliation(s)
| | - Mary-Anne Enoch
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda MD
| | - Colin A. Hodgkinson
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda MD
| | - Marta Pecina
- Molecular and Behavioral Neuroscience Institute, Ann Arbor
| | - Brian Mickey
- Molecular and Behavioral Neuroscience Institute, Ann Arbor,Department of Psychiatry, University of Michigan, Ann Arbor
| | | | | | - David Goldman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda MD
| | - Jon-Kar Zubieta
- Molecular and Behavioral Neuroscience Institute, Ann Arbor,Department of Psychiatry, University of Michigan, Ann Arbor
| |
Collapse
|
38
|
Suridjan I, Boileau I, Bagby M, Rusjan PM, Wilson AA, Houle S, Mizrahi R. Dopamine response to psychosocial stress in humans and its relationship to individual differences in personality traits. J Psychiatr Res 2012; 46:890-7. [PMID: 22475321 DOI: 10.1016/j.jpsychires.2012.03.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2011] [Revised: 03/06/2012] [Accepted: 03/08/2012] [Indexed: 01/27/2023]
Abstract
BACKGROUND Previous studies have reported inter-individual variability in the dopamine (DA) response to stress. This variability might be related to individual differences in the vulnerability to experience the negative effect of stress. OBJECTIVE To investigate whether personality traits as measured by the revised NEO personality inventory explain variability in DA response to a psychosocial stress task. METHODS Eleven healthy adults, mean age of 26 ± 3.87 underwent two positron emission tomography (PET) scans using the dopamine D(2/3) agonist, [11C]-(+)-PHNO under a control and stress condition. The simplified reference tissue model (SRTM) was used to obtain [11C]-(+)-PHNO binding potential (BP(ND)). Stress-induced DA response was indexed as a percent change in [11C]-(+)-PHNO BP(ND) between control and stress conditions. The regions of interest were defined into D2-rich regions, which included the Associative and Sensorimotor Striatum (AST and SMST); D(2/3) mixed regions, which included the limbic striatum (LST) and globus pallidus (GP); and D3-rich region, which included the Substantia Nigra (SN). RESULTS Several personality traits within the Neuroticism and Openness to Experience domain were significantly correlated with blunted DA response to stress. Specifically, the Angry-Hostility, Vulnerability, and Depression trait were associated with blunted DA stress response in the AST (r = -0.645, p = 0.032), LST (r = -0.677, p = 0.022) and GP (r = -0.736, p = 0.010), respectively. The Openness to Values was correlated with a decreased DA release in the SN (r = -0.706, p = 0.015). CONCLUSION Variability in DA stress response might be related to individual differences in personality.
Collapse
Affiliation(s)
- Ivonne Suridjan
- Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Canada
| | | | | | | | | | | | | |
Collapse
|
39
|
te Beek ET, Zoethout RWM, Bani MSG, Andorn A, Iavarone L, Klaassen ES, Fina P, van Gerven JMA. Pharmacokinetics and central nervous system effects of the novel dopamine D3 receptor antagonist GSK598809 and intravenous alcohol infusion at pseudo-steady state. J Psychopharmacol 2012; 26:303-14. [PMID: 22219221 DOI: 10.1177/0269881111431750] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
GSK598809 is a novel selective dopamine D(3) receptor antagonist, currently in development for the treatment of substance abuse and addiction. In a blinded, randomized, placebo-controlled study, effects of single oral doses of 175 mg GSK598809 were evaluated in healthy volunteers. Pharmacokinetics, central nervous system (CNS) effects and potential for interactions with alcohol were evaluated, using an alcohol infusion paradigm and analysis of eye movements, adaptive tracking, visual analogue scales, body sway, serum prolactin and verbal visual learning test. Adverse effects of GSK598809 included headache, dizziness and somnolence. Plasma concentration of GSK598809 was maximal 2-3 hours postdose and decreased with a half-life of roughly 20 hours. CNS effects were limited to prolactin elevation and decreased adaptive tracking. Co-administration of GSK598809 and alcohol did not affect alcohol pharmacokinetics, but caused a 9% decrease of C (max) and a 15% increase of AUC of GSK598809. CNS effects of co-administration were mainly additive, except a small supra-additive increase in saccadic reaction time and decrease in delayed word recall. In conclusion, GSK598809 causes elevation of serum prolactin and a small decrease in adaptive tracking performance. After co-administration with alcohol, effects of GSK598809 are mainly additive and the combination is well tolerated in healthy volunteers.
Collapse
Affiliation(s)
- E T te Beek
- Centre for Human Drug Research, Leiden, the Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Li Y, Kuzhikandathil EV. Molecular characterization of individual D3 dopamine receptor-expressing cells isolated from multiple brain regions of a novel mouse model. Brain Struct Funct 2012; 217:809-33. [PMID: 22286951 DOI: 10.1007/s00429-012-0383-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 01/11/2012] [Indexed: 01/12/2023]
Abstract
Among dopamine receptors, the expression and function of the D3 receptor subtype is not well understood. The receptor has the highest affinity for dopamine and many drugs that target dopamine receptors.In this paper, we examined, at the single cell level, the characteristics of D3 receptor-expressing cells isolated from different brain regions of male and female mice that were either 35 or 70 days old. The brain regions included nucleus accumbens, Islands of Calleja, olfactory tubercle,retrosplenial cortex, dorsal subiculum, mammillary body,amygdala and septum. The expression analysis was done in the drd3-enhanced green fluorescent protein transgenic mice that report the endogenous expression of D3 receptor mRNA. Using single cell reverse transcriptase PCR, we determined if the D3 receptor-expressing fluorescent cells in these mice were neurons or glia and if they were glutamatergic, GABAergic or catecholaminergic. Next, we determined if the fluorescent cells co-expressed the four other dopamine receptor subtypes, adenylate cyclase V(ACV) isoform, and three different isoforms of G protein coupled inward rectifier potassium (GIRK) channels. The results suggest that D3 receptor is expressed in neurons,with region-specific expression in glutamatergic and GABAergic neurons. The D3 receptor primarily coexpressed with D1 and D2 dopamine receptors with regional, sex and age-dependent differences in the coexpression pattern. The percentage of cells co-expressing D3 receptor and ACV or GIRK channels varied significantly by brain region, sex and age. The molecular characterization of D3 receptor-expressing cells in mouse brain reported here will facilitate the characterization of D(3) receptor function in physiology and pathophysiology.
Collapse
Affiliation(s)
- Ying Li
- Department of Pharmacology and Physiology, UMDNJ-New Jersey Medical Schoool, South Orange Avenue, Newark, NJ 07103, USA
| | | |
Collapse
|
41
|
Inta D, Meyer-Lindenberg A, Gass P. Alterations in postnatal neurogenesis and dopamine dysregulation in schizophrenia: a hypothesis. Schizophr Bull 2011; 37:674-80. [PMID: 21097511 PMCID: PMC3122276 DOI: 10.1093/schbul/sbq134] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
An increasing number of studies demonstrate the important role of several susceptibility genes for schizophrenia, such as neuregulin-1 and DISC1, in early postnatal and adult neurogenesis. Its significance for the pathophysiology of the disease, including its relation to neurotransmitter systems implicated in schizophrenia (like the dopamine system), remains, however, unknown. Here, we review molecular and cellular components of the dopamine system associated with postnatal neurogenesis and plasticity, both in rodents and in primates, and discuss their possible implication in schizophrenia. We focus mainly on the islands of Calleja, complex aggregations of granule cells in the ventral striatum, generated early postnatally in the subventricular zone. In contrast to the involution of the primate olfactory bulb, the islands of Calleja attain their maximal development in humans, an evolution paralleled by a larger ventral subventricular zone and more connections with other structures, including temporal cortical areas. The islands of Calleja express high levels of neuronal nitric oxide (NO) synthase and D3 dopamine receptors and are densely interconnected by dopaminergic projections with the ventral tegmental area. D3 receptors modulate subventricular zone neurogenesis and dopamine release. Their genetic deletion induces striatal hyperdopaminergia. We review data indicating a high plasticity of postnatal islands of Calleja, potentially facilitating susceptibility to schizophrenia-related risk factors. In this context, we propose a new pathophysiological model, where altered neurogenesis of the islands of Calleja may contribute to dysfunction of the dopamine and NO systems and psychosis through convergence of genetic and environmental disease-associated factors.
Collapse
Affiliation(s)
- Dragos Inta
- Department for Psychiatry and Psychotherapy, Central Institute of Mental Health Mannheim, University of Heidelberg, Mannheim, Germany.
| | | | | |
Collapse
|
42
|
de Jesús Aceves J, Rueda-Orozco PE, Hernández R, Plata V, Ibañez-Sandoval O, Galarraga E, Bargas J. Dopaminergic presynaptic modulation of nigral afferents: its role in the generation of recurrent bursting in substantia nigra pars reticulata neurons. Front Syst Neurosci 2011; 5:6. [PMID: 21347219 PMCID: PMC3039203 DOI: 10.3389/fnsys.2011.00006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 01/23/2011] [Indexed: 11/13/2022] Open
Abstract
Previous work has shown the functions associated with activation of dopamine presynaptic receptors in some substantia nigra pars reticulata (SNr) afferents: (i) striatonigral terminals (direct pathway) posses presynaptic dopamine D1-class receptors whose action is to enhance inhibitory postsynaptic currents (IPSCs) and GABA transmission. (ii) Subthalamonigral terminals posses D1- and D2-class receptors where D1-class receptor activation enhances and D2-class receptor activation decreases excitatory postsynaptic currents. Here we report that pallidonigral afferents posses D2-class receptors (D3 and D4 types) that decrease inhibitory synaptic transmission via presynaptic modulation. No action of D1-class agonists was found on pallidonigral synapses. In contrast, administration of D1-receptor antagonists greatly decreased striatonigral IPSCs in the same preparation, suggesting that tonic dopamine levels help in maintaining the function of the striatonigral (direct) pathway. When both D3 and D4 type receptors were blocked, pallidonigral IPSCs increased in amplitude while striatonigral connections had no significant change, suggesting that tonic dopamine levels are repressing a powerful inhibition conveyed by pallidonigral synapses (a branch of the indirect pathway). We then blocked both D1- and D2-class receptors to acutely decrease direct pathway (striatonigral) and enhance indirect pathways (subthalamonigral and pallidonigral) synaptic force. The result was that most SNr projection neurons entered a recurrent bursting firing mode similar to that observed during Parkinsonism in both patients and animal models. These results raise the question as to whether the lack of dopamine in basal ganglia output nuclei is enough to generate some pathological signs of Parkinsonism.
Collapse
Affiliation(s)
- José de Jesús Aceves
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México Distrito Federal México, México
| | | | | | | | | | | | | |
Collapse
|
43
|
Searle G, Beaver JD, Comley RA, Bani M, Tziortzi A, Slifstein M, Mugnaini M, Griffante C, Wilson AA, Merlo-Pich E, Houle S, Gunn R, Rabiner EA, Laruelle M. Imaging dopamine D3 receptors in the human brain with positron emission tomography, [11C]PHNO, and a selective D3 receptor antagonist. Biol Psychiatry 2010; 68:392-9. [PMID: 20599188 DOI: 10.1016/j.biopsych.2010.04.038] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Revised: 04/01/2010] [Accepted: 04/01/2010] [Indexed: 10/19/2022]
Abstract
BACKGROUND Dopamine D(3) receptors are involved in the pathophysiology of several neuropsychiatric conditions. [(11)C]-(+)-PHNO is a radiolabeled D(2) and D(3) agonist, suitable for imaging the agonist binding sites (denoted D(2HIGH) and D(3)) of these receptors with positron emission tomography (PET). PET studies in nonhuman primates documented that, in vivo, [(11)C]-(+)-PHNO displays a relative selectivity for D(3) compared with D(2HIGH) receptor sites and that the [(11)C]-(+)-PHNO signal is enriched in D(3) contribution compared with conventional ligands such as [(11)C] raclopride. METHODS To define the D(3) contribution (f(PHNO)(D3)) to [(11)C]-(+)-PHNO binding potential (BP(ND)) in healthy humans, 52 PET scans were obtained in 19 healthy volunteers at baseline and following oral administration of various doses of the selective D(3) receptor antagonist, GSK598809. RESULTS The impact of GSK598809 on [(11)C]-(+)-PHNO was regionally selective. In dorsal regions of the striatum, GSK598809 did not significantly affect [(11)C]-(+)-PHNO BP(ND) (f(PHNO)(D3) approximately 0%). Conversely, in the substantia nigra, GSK598809 dose-dependently reduced [(11)C]-(+)-PHNO binding to nonspecific level (f(PHNO)(D3) approximately 100%). In ventral striatum (VST), globus pallidus and thalamus (THA), [(11)C]-(+)-PHNO BP(ND) was attributable to a combination of D(2HIGH) and D(3) receptor sites, with f(PHNO)(D3) of 26%, 67% and 46%, respectively. D(3) receptor binding potential (BP(ND)(D3)) was highest in globus pallidus (1.90) and substantial nigra (1.39), with lower levels in VST (.77) and THA (.18) and negligible levels in dorsal striatum. CONCLUSIONS This study elucidated the pharmacologic nature of the [(11)C]-(+)-PHNO signal in healthy subjects and provided the first quantification of D(3) receptor availability with PET in the living human brain.
Collapse
Affiliation(s)
- Graham Searle
- Clinical Imaging Centre, GlaxoSmithKline, London, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Hocke C, Maschauer S, Hübner H, Löber S, Utz W, Kuwert T, Gmeiner P, Prante O. A Series of 18F-Labelled Pyridinylphenyl Amides as Subtype-Selective Radioligands for the Dopamine D3 Receptor. ChemMedChem 2010; 5:941-8. [DOI: 10.1002/cmdc.201000067] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
45
|
Rabiner EA, Slifstein M, Nobrega J, Plisson C, Huiban M, Raymond R, Diwan M, Wilson AA, McCormick P, Gentile G, Gunn RN, Laruelle MA. In vivo quantification of regional dopamine-D3 receptor binding potential of (+)-PHNO: Studies in non-human primates and transgenic mice. Synapse 2009; 63:782-93. [PMID: 19489048 DOI: 10.1002/syn.20658] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Examination of dopamine-D3 (D3) receptors with positron emission tomography (PET) have been hampered in the past by the lack of a PET ligand with sufficient selectivity for D3 over dopamine-D2 (D2) receptors. The two types co-localize in the brain, with D2 density significantly higher than D3, hence nonselective PET ligands inform on D2, rather than D3 status. [(11)C]-(+)-PHNO is a novel PET ligand with a preferential affinity for D3 over D2. We used the selective D3 antagonist, SB-277011 to dissect regional fractions of the [(11)C]-(+)-PHNO signal attributable to D3 and D2 in primate brain. The results were compared with quantitative autoradiography with (3)H-(+)-PHNO in wild-type, D2-knock-out, and D3-knock-out mice examined at baseline and following administration of SB-277011. Both sets of results converged to indicate a predominant D3-related component to (+)-PHNO binding in extra-striatal regions, with binding in the midbrain being entirely attributable to D3. The midbrain is thus an excellent target region to examine D3 receptor occupancy with [(11)C]-(+)-PHNO PET in vivo.
Collapse
Affiliation(s)
- Eugenii A Rabiner
- GlaxoSmithKline Clinical Imaging Centre, Hammersmith Hospital, London, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Seeman P, Guan HC. Glutamate agonists for treating schizophrenia have affinity for dopamine D2Highand D3 receptors. Synapse 2009; 63:705-9. [DOI: 10.1002/syn.20673] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
47
|
Seeman P. Dopamine D2High receptors measured ex vivo are elevated in amphetamine-sensitized animals. Synapse 2009; 63:186-92. [PMID: 19086090 DOI: 10.1002/syn.20595] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Although dopamine supersensitivity is a fundamental aspect of diseases such as schizophrenia and Parkinson's disease, the molecular basis of dopamine supersensitivity is not known. Because behavioral dopamine supersensitivity is associated with a marked elevation of striatal dopamine D2(High) receptors in vitro, it is important to develop methods to measure D2(High) receptors in vivo. The present ex vivo study found that the dopamine agonist NPA ([-]-N-propyl-norapomorphine) inhibited the binding of the agonist [(3)H](+)PHNO to rat striatal D2 receptors significantly more than the D2 antagonist [(3)H]raclopride, when NPA was coinjected i.v. with each radioligand. These results suggest that the greater sensitivity of [(3)H](+)PHNO to inhibition by the coinjected NPA reflects in vivo competition at D2(High) receptors. Using rats that had been sensitized to amphetamine, this ex vivo method found that the specific binding of [(3)H](+)PHNO that was displaced by 10 microg/kg of NPA was 2.4-fold higher than that for control rats. These data agree with in vitro data showing a marked increase in D2(High) sites after amphetamine sensitization. Therefore, it is recommended that this method of co-injecting the D2 radioligand and the dopamine agonist displacer be used in human positron tomography to detect D2(High) receptors in health and disease.
Collapse
Affiliation(s)
- Philip Seeman
- Clera Inc., 260 Heath Street West, Toronto, Ontario, Canada.
| |
Collapse
|
48
|
Seeman P, Battaglia G, Corti C, Corsi M, Bruno V. Glutamate receptor mGlu2 and mGlu3 knockout striata are dopamine supersensitive, with elevated D2(High) receptors and marked supersensitivity to the dopamine agonist (+)PHNO. Synapse 2009; 63:247-51. [PMID: 19084908 DOI: 10.1002/syn.20607] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The finding that the mGlu2/3 metabotropic glutamate receptor agonist, LY404039, improves clinical symptoms in schizophrenia warrants a search for a possible interaction between mGlu2/3 receptors and dopamine D2 receptors. Here, this topic is examined in striatal tissue of mice lacking either mGlu2 or mGlu3 receptor. Such mice are known to be behaviorally supersensitive to dopamine receptor agonists. Therefore, to determine the basis of this dopamine supersensitivity, the proportion of dopamine D2(High) receptors was measured in the striata of mGlu2 and mGlu3 receptor knockout mice. The proportion of D2(High) receptors was found to be elevated by 220% in the striata of both knockouts. To measure the functional dopamine supersensitivity, the D2 agonist (+)PHNO was used to stimulate the incorporation of GTP-gamma-S in the striatal homogenates in the presence of drugs that blocked the dopamine D1, D3, and D5 receptors. Compared with control striata, the mGlu2 receptor knockout tissues were 67-fold more sensitive to (+)PHNO, while the mGlu3 receptor knockout tissues were 17-fold more sensitive. These data suggest that group II mGlu receptors-mGlu2 receptors in particular-may normally regulate D2 receptors by reducing the proportion of high-affinity D2 receptors in membranes. Such regulation may contribute to the antipsychotic action of mGlu2/3 receptor agonists.
Collapse
Affiliation(s)
- Philip Seeman
- Department of Pharmacology, Medical Science Building, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada.
| | | | | | | | | |
Collapse
|
49
|
Abstract
Expectations, positive or negative, are modulating factors influencing behavior. They are also thought to underlie placebo effects, potentially impacting perceptions and biological processes. We used sustained pain as a model to determine the neural mechanisms underlying placebo-induced analgesia and affective changes in healthy humans. Subjects were informed that they could receive either an active agent or an inactive compound, similar to routine clinical trials. Using PET and the mu-opioid selective radiotracer [(11)C]carfentanil we demonstrate placebo-induced activation of opioid neurotransmission in a number of brain regions. These include the rostral anterior cingulate, orbitofrontal and dorsolateral prefrontal cortex, anterior and posterior insula, nucleus accumbens, amygdala, thalamus, hypothalamus, and periaqueductal grey. Some of these regions overlap with those involved in pain and affective regulation but also motivated behavior. The activation of endogenous opioid neurotransmission was further associated with reductions in pain report and negative affective state. Additional studies with the radiotracer [(11)C]raclopride, studies labeling dopamine D2/3 receptors, also demonstrate the activation of nucleus accumbens dopamine during placebo administration under expectation of analgesia. Both dopamine and opioid neurotransmission were related to expectations of analgesia and deviations from those initial expectations. When the activity of the nucleus accumbens was probed with fMRI using a monetary reward expectation paradigm, its activation was correlated with both dopamine, opioid responses to placebo in this region and the formation of placebo analgesia. These data confirm that specific neural circuits and neurotransmitter systems respond to the expectation of benefit during placebo administration, inducing measurable physiological changes.
Collapse
Affiliation(s)
- Jon-Kar Zubieta
- Department of Psychiatry and Molecular and Behavioral Neuroscience Institute, University of Maryland, Baltimore, USA.
| | | |
Collapse
|
50
|
Boileau I, Guttman M, Rusjan P, Adams JR, Houle S, Tong J, Hornykiewicz O, Furukawa Y, Wilson AA, Kapur S, Kish SJ. Decreased binding of the D3 dopamine receptor-preferring ligand [11C]-(+)-PHNO in drug-naive Parkinson's disease. Brain 2009; 132:1366-75. [PMID: 19153147 DOI: 10.1093/brain/awn337] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The D(3) dopamine (DA) receptor is a member of the D(2)-like DA receptor family. While the D(2) receptor is abundant especially in motor-regions of the striatum, the D(3) receptor shows a relative abundance in limbic regions and globus pallidus. This receptor is of current interest in neurology because of its potential involvement in psychiatric and motor complications in Parkinson's disease and the possibility that dopamine D(3)-preferring agonist therapy might delay progression of the disorder. Preclinical data indicate that striatal levels of the D(3) (but not the D(2)) DA receptor are decreased following lesion of nigrostriatal DA neurons; at present, there are no in vivo data on this receptor subtype in Parkinson's disease. The objective of this positron emission tomography study was to compare [(11)C]-(+)-PHNO (D(3) versus D(2) preferring) and [(11)C]raclopride (D(3) = D(2)) binding in brain of non-depressed, non-demented, dopaminergic drug-naïve patients with early-stage Parkinson's disease (n = 10), relative to matched-controls (n = 9). Parkinson's disease was associated with a trend for bilaterally decreased [(11)C]-(+)-PHNO (but not [(11)C]raclopride) binding in the D(3)-rich ventral striatum (-11%, P = 0.07) and significantly decreased binding in globus pallidus (-42%, P = 0.02). In contrast, in the primarily D(2)-populated putamen, both [(11)C]-(+)-PHNO (25%, P = 0.02) and [(11)C]raclopride (25%, P < 0.01) binding were similarly increased, especially on the side contra-lateral to the symptoms. In the midbrain, presumably containing D(3) receptors localized to the substantia nigra, [(11)C]-(+)-PHNO binding was normal. Decreased [(11)C]-(+)-PHNO to [(11)C]raclopride ratio correlated with motor deficits and lowered-mood (P < 0.02). Our imaging data suggest that brain DA neuron loss in the human causes region-specific differential changes in DA D(2) and D(3) receptors with D(3) receptor 'downregulation' possibly related to some motor and mood problems in Parkinson disease. D(3) receptor levels might be a determinant vulnerability factor underlying side-effects associated with treatment; hence, these initial findings provide valuable baseline information to understand the role of D(3) receptors in response to Parkinson's disease medication.
Collapse
Affiliation(s)
- Isabelle Boileau
- Human Neurochemical Pathology Laboratory, University of Toronto, Toronto, Ontario, Canada.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|