1
|
Saoud H, Foddha H, Aflouk Y, Jrad BBH. Involvement of CXCL10 rs4256246, CXCR4 rs2228014, CCR2 rs1799864 and CXCL16 rs2277680 in the Predisposition to Schizophrenia. J Mol Neurosci 2024; 74:86. [PMID: 39264476 DOI: 10.1007/s12031-024-02257-9] [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: 03/21/2024] [Accepted: 08/14/2024] [Indexed: 09/13/2024]
Abstract
Chemokine ligands and their receptors have acquired less attention than pro- and anti-inflammatory cytokines in schizophrenia (SCZ). Thus, we aimed to examine the impact of functional polymorphisms of the chemokine genes CXCL10, CXCL16, CXCR4, and CCR2 in the development of SCZ. Using PCR-RFLP, we analyzed the selected polymorphisms in a Tunisian cohort composed of 200 patients with SCZ and 200 healthy controls. Our preliminary data suggest that the minor allele A of CXCL10 rs4256246 is significantly associated with likelihood of SCZ (PAdjusted = 0.00002) and more precisely to paranoid patients with late-onset SCZ (PAdjusted = 0.0007). However, the mutated allele T of CXCR4 rs2228014 showed a significant protective impact against SCZ (PAdjusted = 0.000007) and especially to male sex (PAdjusted = 0.000003). This effect persists among the undifferentiated patients with early-onset SCZ (PAdjusted = 0.002). Following the stratified analyses, CCR2 rs1799864 and CXCL16 rs2277680 were significantly correlated with the clinical symptoms among disorganized patients. As regards haplotype analysis, we noted that GATG haplotype was associated with protection against SCZ (PAdjusted = 0.0087) but the AGCG haplotype was correlated with susceptibility to this disease (PAdjusted = 0.014). Our preliminary results suggested that CXCL10 rs4256246 enhanced susceptibility to SCZ, while CXCR4 rs2228014 seemed to be protective factor. Furthermore, we identified a substantial correlation between CCR2 rs1799864 and CXCL16 rs2277680 with the clinical signs of the disorder. To validate these results and clarify the functional significance of the targeted polymorphisms in SCZ, more independent research is needed.
Collapse
Affiliation(s)
- Hana Saoud
- Laboratory of Genetics, Biodiversity and Bioresource Valorization (LR11ES41), Higher Institute of Biotechnology of Monastir, University of Monastir, Monastir, Tunisia.
| | - Hajer Foddha
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Youssef Aflouk
- Laboratory of Genetics, Biodiversity and Bioresource Valorization (LR11ES41), Higher Institute of Biotechnology of Monastir, University of Monastir, Monastir, Tunisia
| | - Besma Bel Hadj Jrad
- Laboratory of Genetics, Biodiversity and Bioresource Valorization (LR11ES41), Higher Institute of Biotechnology of Monastir, University of Monastir, Monastir, Tunisia
| |
Collapse
|
2
|
Carmichael O. The Role of fMRI in Drug Development: An Update. ADVANCES IN NEUROBIOLOGY 2023; 30:299-333. [PMID: 36928856 DOI: 10.1007/978-3-031-21054-9_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Functional magnetic resonance imaging (fMRI) of the brain is a technology that holds great potential for increasing the efficiency of drug development for the central nervous system (CNS). In preclinical studies and both early- and late-phase human trials, fMRI has the potential to improve cross-species translation of drug effects, help to de-risk compounds early in development, and contribute to the portfolio of evidence for a compound's efficacy and mechanism of action. However, to date, the utilization of fMRI in the CNS drug development process has been limited. The purpose of this chapter is to explore this mismatch between potential and utilization. This chapter provides introductory material related to fMRI and drug development, describes what is required of fMRI measurements for them to be useful in a drug development setting, lists current capabilities of fMRI in this setting and challenges faced in its utilization, and ends with directions for future development of capabilities in this arena. This chapter is the 5-year update of material from a previously published workshop summary (Carmichael et al., Drug DiscovToday 23(2):333-348, 2018).
Collapse
Affiliation(s)
- Owen Carmichael
- Pennington Biomedical Research Center, Baton Rouge, LA, USA.
| |
Collapse
|
3
|
Gärtner M, de Rover M, Václavů L, Scheidegger M, van Osch MJP, Grimm S. Increase in thalamic cerebral blood flow is associated with antidepressant effects of ketamine in major depressive disorder. World J Biol Psychiatry 2022; 23:643-652. [PMID: 34985394 DOI: 10.1080/15622975.2021.2020900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Ketamine is a promising treatment option for patients with Major Depressive Disorder (MDD) and has become an important research tool to investigate antidepressant mechanisms of action. However, imaging studies attempting to characterise ketamine's mechanism of action using blood oxygen level-dependent signal (BOLD) imaging have yielded inconsistent results- at least partly due to intrinsic properties of the BOLD contrast, which measures a complex signal related to neural activity. To circumvent the limitations associated with the BOLD signal, we used arterial spin labelling (ASL) as an unambiguous marker of neuronal activity-related changes in cerebral blood flow (CBF). We measured CBF in 21 MDD patients at baseline and 24 h after receiving a single intravenous infusion of subanesthetic ketamine and examined relationships with clinical outcomes. Our findings demonstrate that increase in thalamus perfusion 24 h after ketamine administration is associated with greater improvement of depressive symptoms. Furthermore, lower thalamus perfusion at baseline is associated both with larger increases in perfusion 24 h after ketamine administration and with stronger reduction of depressive symptoms. These findings indicate that ASL is not only a useful tool to broaden our understanding of ketamine's mechanism of action but might also have the potential to inform treatment decisions based on CBF-defined regional disruptions.
Collapse
Affiliation(s)
- Matti Gärtner
- MSB-Medical School Berlin, Berlin, Germany.,Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Mischa de Rover
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands.,Department of Clinical Psychology, Institute of Psychology, Leiden University, Leiden, Netherlands
| | - Lena Václavů
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, Netherlands
| | - Milan Scheidegger
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| | - Matthias J P van Osch
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, Netherlands
| | - Simone Grimm
- MSB-Medical School Berlin, Berlin, Germany.,Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| |
Collapse
|
4
|
The M1/M4 preferring muscarinic agonist xanomeline modulates functional connectivity and NMDAR antagonist-induced changes in the mouse brain. Neuropsychopharmacology 2021; 46:1194-1206. [PMID: 33342996 PMCID: PMC8115158 DOI: 10.1038/s41386-020-00916-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/02/2020] [Accepted: 11/10/2020] [Indexed: 12/25/2022]
Abstract
Cholinergic drugs acting at M1/M4 muscarinic receptors hold promise for the treatment of symptoms associated with brain disorders characterized by cognitive impairment, mood disturbances, or psychosis, such as Alzheimer's disease or schizophrenia. However, the brain-wide functional substrates engaged by muscarinic agonists remain poorly understood. Here we used a combination of pharmacological fMRI (phMRI), resting-state fMRI (rsfMRI), and resting-state quantitative EEG (qEEG) to investigate the effects of a behaviorally active dose of the M1/M4-preferring muscarinic agonist xanomeline on brain functional activity in the rodent brain. We investigated both the effects of xanomeline per se and its modulatory effects on signals elicited by the NMDA-receptor antagonists phencyclidine (PCP) and ketamine. We found that xanomeline induces robust and widespread BOLD signal phMRI amplitude increases and decreased high-frequency qEEG spectral activity. rsfMRI mapping in the mouse revealed that xanomeline robustly decreased neocortical and striatal connectivity but induces focal increases in functional connectivity within the nucleus accumbens and basal forebrain. Notably, xanomeline pre-administration robustly attenuated both the cortico-limbic phMRI response and the fronto-hippocampal hyper-connectivity induced by PCP, enhanced PCP-modulated functional connectivity locally within the nucleus accumbens and basal forebrain, and reversed the gamma and high-frequency qEEG power increases induced by ketamine. Collectively, these results show that xanomeline robustly induces both cholinergic-like neocortical activation and desynchronization of functional networks in the mammalian brain. These effects could serve as a translatable biomarker for future clinical investigations of muscarinic agents, and bear mechanistic relevance for the putative therapeutic effect of these class of compounds in brain disorders.
Collapse
|
5
|
Mervin LH, Mitricheva E, Logothetis NK, Bifone A, Bender A, Noori HR. Neurochemical underpinning of hemodynamic response to neuropsychiatric drugs: A meta- and cluster analysis of preclinical studies. J Cereb Blood Flow Metab 2021; 41:874-885. [PMID: 32281457 PMCID: PMC7983335 DOI: 10.1177/0271678x20916003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/26/2020] [Accepted: 03/04/2020] [Indexed: 11/30/2022]
Abstract
Functional magnetic resonance imaging (fMRI) is an extensively used method for the investigation of normal and pathological brain function. In particular, fMRI has been used to characterize spatiotemporal hemodynamic response to pharmacological challenges as a non-invasive readout of neuronal activity. However, the mechanisms underlying regional signal changes are yet unclear. In this study, we use a meta-analytic approach to converge data from microdialysis experiments with relative cerebral blood volume (rCBV) changes following acute administration of neuropsychiatric drugs in adult male rats. At whole-brain level, the functional response patterns show very weak correlation with neurochemical alterations, while for numerous brain areas a strong positive correlation with noradrenaline release exists. At a local scale of individual brain regions, the rCBV response to neurotransmitters is anatomically heterogeneous and, importantly, based on a complex interplay of different neurotransmitters that often exert opposing effects, thus providing a mechanism for regulating and fine tuning hemodynamic responses in specific regions.
Collapse
Affiliation(s)
- Lewis H Mervin
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Ekaterina Mitricheva
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Nikos K Logothetis
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
- Imaging Science and Biomedical Engineering, University of Manchester, Manchester, UK
| | - Angelo Bifone
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, Rovereto, Italy
| | - Andreas Bender
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Hamid R Noori
- Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
6
|
Kantrowitz JT, Grinband J, Goff DC, Lahti AC, Marder SR, Kegeles LS, Girgis RR, Sobeih T, Wall MM, Choo TH, Green MF, Yang YS, Lee J, Horga G, Krystal JH, Potter WZ, Javitt DC, Lieberman JA. Proof of mechanism and target engagement of glutamatergic drugs for the treatment of schizophrenia: RCTs of pomaglumetad and TS-134 on ketamine-induced psychotic symptoms and pharmacoBOLD in healthy volunteers. Neuropsychopharmacology 2020; 45:1842-1850. [PMID: 32403118 PMCID: PMC7608251 DOI: 10.1038/s41386-020-0706-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/12/2020] [Accepted: 05/06/2020] [Indexed: 12/15/2022]
Abstract
Glutamate neurotransmission is a prioritized target for antipsychotic drug development. Two metabotropic glutamate receptor 2/3 (mGluR2/3) agonists (pomaglumetad [POMA] and TS-134) were assessed in two Phase Ib proof of mechanism studies of comparable designs and using identical clinical assessments and pharmacoBOLD methodology. POMA was examined in a randomized controlled trial under double-blind conditions for 10-days at doses of 80 or 320 mg/d POMA versus placebo (1:1:1 ratio). The TS-134 trial was a randomized, single-blind, 6-day study of 20 or 60 mg/d TS-134 versus placebo (5:5:2 ratio). Primary outcomes were ketamine-induced changes in pharmacoBOLD in the dorsal anterior cingulate cortex (dACC) and symptoms reflected on the Brief Psychiatric Rating Scale (BPRS). Both trials were conducted contemporaneously. 95 healthy volunteers were randomized to POMA and 63 to TS-134. High-dose POMA significantly reduced ketamine-induced BPRS total symptoms within and between-groups (p < 0.01, d = -0.41; p = 0.04, d = -0.44, respectively), but neither POMA dose significantly suppressed ketamine-induced dACC pharmacoBOLD. In contrast, low-dose TS-134 led to moderate to large within and between group reductions in both BPRS positive symptoms (p = 0.02, d = -0.36; p = 0.008, d = -0.82, respectively) and dACC pharmacoBOLD (p = 0.004, d = -0.56; p = 0.079, d = -0.50, respectively) using pooled across-study placebo data. High-dose POMA exerted significant effects on clinical symptoms, but not on target engagement, suggesting a higher dose may yet be needed, while the low dose of TS-134 showed evidence of symptom reduction and target engagement. These results support further investigation of mGluR2/3 and other glutamate-targeted treatments for schizophrenia.
Collapse
Affiliation(s)
- Joshua T. Kantrowitz
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA ,grid.250263.00000 0001 2189 4777Nathan Kline Institute, Orangeburg, NY USA
| | - Jack Grinband
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| | - Donald C. Goff
- grid.250263.00000 0001 2189 4777Nathan Kline Institute, Orangeburg, NY USA ,grid.240324.30000 0001 2109 4251NYU Langone Medical Center, New York, NY USA
| | - Adrienne C. Lahti
- grid.265892.20000000106344187University of Alabama at Birmingham, Birmingham, AL USA
| | | | - Lawrence S. Kegeles
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| | - Ragy R. Girgis
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| | - Tarek Sobeih
- grid.250263.00000 0001 2189 4777Nathan Kline Institute, Orangeburg, NY USA
| | - Melanie M. Wall
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| | - Tse-Hwei Choo
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| | | | - Yvonne S. Yang
- grid.19006.3e0000 0000 9632 6718UCLA, Los Angeles, CA USA
| | - Junghee Lee
- grid.19006.3e0000 0000 9632 6718UCLA, Los Angeles, CA USA
| | - Guillermo Horga
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| | - John H. Krystal
- grid.47100.320000000419368710Yale University School of Medicine, New Haven, CT USA
| | - William Z. Potter
- grid.94365.3d0000 0001 2297 5165National Institutes of Health, Bethesda, MD USA
| | - Daniel C. Javitt
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA ,grid.250263.00000 0001 2189 4777Nathan Kline Institute, Orangeburg, NY USA
| | - Jeffrey A. Lieberman
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| |
Collapse
|
7
|
Watanabe M, Marcy B, Kinoshita K, Fukasawa M, Hikichi H, Chaki S, Okuyama S, Gevorkyan H, Yoshida S. Safety and pharmacokinetic profiles of MGS0274 besylate (TS-134), a novel metabotropic glutamate 2/3 receptor agonist prodrug, in healthy subjects. Br J Clin Pharmacol 2020; 86:2286-2301. [PMID: 32353162 PMCID: PMC7576618 DOI: 10.1111/bcp.14331] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/30/2020] [Accepted: 04/20/2020] [Indexed: 01/07/2023] Open
Abstract
Aims The safety and pharmacokinetics of single and multiple doses of a novel mGlu2/3 receptor agonist prodrug, MGS0274 besylate (TS‐134), were investigated in healthy subjects. Methods Phase 1 single‐ascending dose (5–20 mg) and multiple‐ascending dose titration (5–80 mg) studies were conducted in healthy male and female subjects. Both studies were randomized, double‐blinded and placebo‐controlled. In one cohort of single‐ascending dose study (10 mg), concentrations of MGS0008, the active compound, in the cerebrospinal fluid (CSF) were measured for up to 24 hours postdose. Results Following single and multiple oral administrations, MGS0274 was rapidly absorbed and extensively converted into MGS0008, which reached a maximum concentration (Cmax) in plasma within 4 hours postdose and declined with a terminal half‐life (t1/2) of around 10 hours. Plasma exposure to MGS0274 was minimal, accounting for approximately 3% of the area under the concentration–time curve (AUC) of MGS0008. Plasma Cmax and AUC of MGS0008 at steady state increased dose proportionally (5–80 mg). MGS0008 penetrated into CSF, with a CSF‐to‐plasma Cmax ratio of 3.66%, and was eliminated with a t1/2 of approximately 16 hours. The most frequent treatment‐emergent adverse events observed following single and multiple oral administration included headache, nausea, somnolence, dizziness and vomiting. Conclusion TS‐134 is orally bioavailable in humans and converts rapidly and extensively to MGS0008, which exhibits good CSF penetration. Orally administered TS‐134 was safe and generally well‐tolerated; hence, TS‐134 is a promising candidate for further clinical development for the treatment of disorders in which glutamatergic abnormalities are involved, such as schizophrenia.
Collapse
|
8
|
Mandino F, Cerri DH, Garin CM, Straathof M, van Tilborg GAF, Chakravarty MM, Dhenain M, Dijkhuizen RM, Gozzi A, Hess A, Keilholz SD, Lerch JP, Shih YYI, Grandjean J. Animal Functional Magnetic Resonance Imaging: Trends and Path Toward Standardization. Front Neuroinform 2020; 13:78. [PMID: 32038217 PMCID: PMC6987455 DOI: 10.3389/fninf.2019.00078] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 12/19/2019] [Indexed: 12/21/2022] Open
Abstract
Animal whole-brain functional magnetic resonance imaging (fMRI) provides a non-invasive window into brain activity. A collection of associated methods aims to replicate observations made in humans and to identify the mechanisms underlying the distributed neuronal activity in the healthy and disordered brain. Animal fMRI studies have developed rapidly over the past years, fueled by the development of resting-state fMRI connectivity and genetically encoded neuromodulatory tools. Yet, comparisons between sites remain hampered by lack of standardization. Recently, we highlighted that mouse resting-state functional connectivity converges across centers, although large discrepancies in sensitivity and specificity remained. Here, we explore past and present trends within the animal fMRI community and highlight critical aspects in study design, data acquisition, and post-processing operations, that may affect the results and influence the comparability between studies. We also suggest practices aimed to promote the adoption of standards within the community and improve between-lab reproducibility. The implementation of standardized animal neuroimaging protocols will facilitate animal population imaging efforts as well as meta-analysis and replication studies, the gold standards in evidence-based science.
Collapse
Affiliation(s)
- Francesca Mandino
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research, Singapore, Singapore
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Domenic H. Cerri
- Center for Animal MRI, Department of Neurology, Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Clement M. Garin
- Direction de la Recherche Fondamentale, MIRCen, Institut de Biologie François Jacob, Commissariat à l’Énergie Atomique et aux Énergies Alternatives, Fontenay-aux-Roses, France
- Neurodegenerative Diseases Laboratory, Centre National de la Recherche Scientifique, UMR 9199, Université Paris-Sud, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Milou Straathof
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Geralda A. F. van Tilborg
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - M. Mallar Chakravarty
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Department of Biological and Biomedical Engineering, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Marc Dhenain
- Direction de la Recherche Fondamentale, MIRCen, Institut de Biologie François Jacob, Commissariat à l’Énergie Atomique et aux Énergies Alternatives, Fontenay-aux-Roses, France
- Neurodegenerative Diseases Laboratory, Centre National de la Recherche Scientifique, UMR 9199, Université Paris-Sud, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Rick M. Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Centre for Neuroscience and Cognitive Systems @ UNITN, Rovereto, Italy
| | - Andreas Hess
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich–Alexander University Erlangen–Nürnberg, Erlangen, Germany
| | - Shella D. Keilholz
- Department of Biomedical Engineering, Georgia Tech, Emory University, Atlanta, GA, United States
| | - Jason P. Lerch
- Hospital for Sick Children, Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Wellcome Centre for Integrative NeuroImaging, University of Oxford, Oxford, United Kingdom
| | - Yen-Yu Ian Shih
- Center for Animal MRI, Department of Neurology, Biomedical Research Imaging Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Joanes Grandjean
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Radiology and Nuclear Medicine, Donders Institute for Brain, Cognition, and Behaviour, Donders Institute, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
9
|
Acute and Repeated Intranasal Oxytocin Differentially Modulate Brain-wide Functional Connectivity. Neuroscience 2020; 445:83-94. [PMID: 31917352 DOI: 10.1016/j.neuroscience.2019.12.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 12/31/2022]
Abstract
Central release of the neuropeptide oxytocin (OXT) modulates neural substrates involved in socio-affective behavior. This property has prompted research into the use of intranasal OXT administration as an adjunctive therapy for brain conditions characterized by social impairment, such as autism spectrum disorders (ASD). However, the neural circuitry and brain-wide functional networks recruited by intranasal OXT administration remain elusive. Moreover, little is known of the neuroadaptive cascade triggered by long-term administration of this peptide at the network level. To address these questions, we applied fMRI-based circuit mapping in adult mice upon acute and repeated (seven-day) intranasal dosing of OXT. We report that acute and chronic OXT administration elicit comparable fMRI activity as assessed with cerebral blood volume mapping, but entail largely different patterns of brain-wide functional connectivity. Specifically, acute OXT administration focally boosted connectivity within key limbic components of the rodent social brain, whereas repeated dosing led to a prominent and widespread increase in functional connectivity, involving a strong coupling between the amygdala and extended cortical territories. Importantly, this connectional reconfiguration was accompanied by a paradoxical reduction in social interaction and communication in wild-type mice. Our results identify the network substrates engaged by exogenous OXT administration, and show that repeated OXT dosing leads to a substantial reconfiguration of brain-wide connectivity, entailing an aberrant functional coupling between cortico-limbic structures involved in socio-communicative and affective functions. Such divergent patterns of network connectivity might contribute to discrepant clinical findings involving acute or long-term OXT dosing in clinical populations.
Collapse
|
10
|
Protective Effect of the MCP-1 Gene Haplotype against Schizophrenia. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4042615. [PMID: 31886209 PMCID: PMC6925699 DOI: 10.1155/2019/4042615] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/28/2019] [Accepted: 11/15/2019] [Indexed: 12/24/2022]
Abstract
While cytokines and their genetic variants have been intensively studied in schizophrenia, little attention has been focused on chemokines in the last years. The monocyte chemoattractant protein 1 (MCP-1) is known to attract peripheral monocytes to the brain during an inflammatory reaction and to affect the T helper (Th) cell development by stimulating Th2 polarization. Owing to the neuroinflammation in schizophrenia and the variable level of MCP-1 in these patients' sera, we proposed to analyze the impact of functional genetic variants of the MCP-1 gene (MCP-1-2518A/G (rs1024611), MCP-1-362G/C (rs2857656), and MCP-1 int1del554-567 (rs3917887)) in schizophrenic patients. We conducted a case-control study on a Tunisian population composed of 200 patients and 200 controls using RFLP-PCR. Our results indicated that the minor alleles (-2518G and Del554-567) were significantly more prevalent in controls than in patients (P=0.001/adjusted OR = 0.42, P=0.04/adjusted OR = 0.64), whereas, for -362C minor allele, increased risk of schizophrenia was revealed (P=0.001, adjusted OR = 2.38). In conclusion, we have identified the haplotype combination -2581G/-362G/int1del554-567 that could mediate protection against schizophrenia (P=0.0038, OR = 0.19) and the effect could result more strongly from the MCP-1 -2582G with -362G variants, whereas the effect of int1del554-567 may in part be explained by its LD with -362.
Collapse
|
11
|
Doostdar N, Kim E, Grayson B, Harte MK, Neill JC, Vernon AC. Global brain volume reductions in a sub-chronic phencyclidine animal model for schizophrenia and their relationship to recognition memory. J Psychopharmacol 2019; 33:1274-1287. [PMID: 31060435 DOI: 10.1177/0269881119844196] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Cognitive deficits and structural brain changes co-occur in patients with schizophrenia. Improving our understanding of the relationship between these is important to develop improved therapeutic strategies. Back-translation of these findings into rodent models for schizophrenia offers a potential means to achieve this goal. AIMS The purpose of this study was to determine the extent of structural brain changes and how these relate to cognitive behaviour in a sub-chronic phencyclidine rat model. METHODS Performance in the novel object recognition task was examined in female Lister Hooded rats at one and six weeks after sub-chronic phencyclidine (2 mg/kg intra-peritoneal, n=15) and saline controls (1 ml/kg intra-peritoneal, n=15). Locomotor activity following acute phencyclidine challenge was also measured. Brain volume changes were assessed in the same animals using ex vivo structural magnetic resonance imaging and computational neuroanatomical analysis at six weeks. RESULTS Female sub-chronic phencyclidine-treated Lister Hooded rats spent significantly less time exploring novel objects (p<0.05) at both time-points and had significantly greater locomotor activity response to an acute phencyclidine challenge (p<0.01) at 3-4 weeks of washout. At six weeks, sub-chronic phencyclidine-treated Lister Hooded rats displayed significant global brain volume reductions (p<0.05; q<0.05), without apparent regional specificity. Relative volumes of the perirhinal cortex however were positively correlated with novel object exploration time only in sub-chronic phencyclidine rats at this time-point. CONCLUSION A sustained sub-chronic phencyclidine-induced cognitive deficit in novel object recognition is accompanied by global brain volume reductions in female Lister Hooded rats. The relative volumes of the perirhinal cortex however are positively correlated with novel object exploration, indicating some functional relevance.
Collapse
Affiliation(s)
- Nazanin Doostdar
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Eugene Kim
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Ben Grayson
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Michael K Harte
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Joanna C Neill
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Anthony C Vernon
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| |
Collapse
|
12
|
( 2R,6R)-hydroxynorketamine exerts mGlu 2 receptor-dependent antidepressant actions. Proc Natl Acad Sci U S A 2019; 116:6441-6450. [PMID: 30867285 DOI: 10.1073/pnas.1819540116] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Currently approved antidepressant drugs often take months to take full effect, and ∼30% of depressed patients remain treatment resistant. In contrast, ketamine, when administered as a single subanesthetic dose, exerts rapid and sustained antidepressant actions. Preclinical studies indicate that the ketamine metabolite (2R,6R)-hydroxynorketamine [(2R,6R)-HNK] is a rapid-acting antidepressant drug candidate with limited dissociation properties and abuse potential. We assessed the role of group II metabotropic glutamate receptor subtypes 2 (mGlu2) and 3 (mGlu3) in the antidepressant-relevant actions of (2R,6R)-HNK using behavioral, genetic, and pharmacological approaches as well as cortical quantitative EEG (qEEG) measurements in mice. Both ketamine and (2R,6R)-HNK prevented mGlu2/3 receptor agonist (LY379268)-induced body temperature increases in mice lacking the Grm3, but not Grm2, gene. This action was not replicated by NMDA receptor antagonists or a chemical variant of ketamine that limits metabolism to (2R,6R)-HNK. The antidepressant-relevant behavioral effects and 30- to 80-Hz qEEG oscillation (gamma-range) increases resultant from (2R,6R)-HNK administration were prevented by pretreatment with an mGlu2/3 receptor agonist and absent in mice lacking the Grm2, but not Grm3 -/-, gene. Combined subeffective doses of the mGlu2/3 receptor antagonist LY341495 and (2R,6R)-HNK exerted synergistic increases on gamma oscillations and antidepressant-relevant behavioral actions. These findings highlight that (2R,6R)-HNK exerts antidepressant-relevant actions via a mechanism converging with mGlu2 receptor signaling and suggest enhanced cortical gamma oscillations as a marker of target engagement relevant to antidepressant efficacy. Moreover, these results support the use of (2R,6R)-HNK and inhibitors of mGlu2 receptor function in clinical trials for treatment-resistant depression either alone or in combination.
Collapse
|
13
|
Bertron JL, Seto M, Lindsley CW. DARK Classics in Chemical Neuroscience: Phencyclidine (PCP). ACS Chem Neurosci 2018; 9:2459-2474. [PMID: 29953199 DOI: 10.1021/acschemneuro.8b00266] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Phencyclidine (PCP, "angel dust", an arylcyclohexylamine) was the first non-natural, man-made illicit drug of abuse, and was coined 'the most dangerous drug in America" in the late 1970s (amidst sensational horror stories of the drug's effects); however, few other illicit drugs have had such a significant and broad impact on society-both good and bad. Originally developed as a new class of anesthetic, PCP-derived psychosis gave way to the PCP hypothesis of schizophrenia (later coined the NMDA receptor hypofunction hypothesis or the glutamate hypothesis of schizophrenia), which continues to drive therapeutic discovery for schizophrenia today. PCP also led to the discovery of ketamine (and a new paradigm for the treatment of major depression), as well as other illicit, designer drugs, such as methoxetamine (MXE) and a new wave of Internet commerce for illicit drugs (sold as research chemicals, or RCs). Furthermore, PCP is a significant contaminant/additive of many illegal drugs sold today, due to its ease of preparation by clandestine chemists. Here, we will review the history, importance, synthesis (both legal and clandestine), pharmacology, drug metabolism, and folklore of PCP, a true DARK classic in chemical neuroscience.
Collapse
Affiliation(s)
- Jeanette L. Bertron
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Chemistry, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
| | - Mabel Seto
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Craig W. Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Chemistry, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
14
|
Mehta MA, Schmechtig A, Kotoula V, McColm J, Jackson K, Brittain C, Tauscher-Wisniewski S, Kinon BJ, Morrison PD, Pollak T, Mant T, Williams SCR, Schwarz AJ. Group II metabotropic glutamate receptor agonist prodrugs LY2979165 and LY2140023 attenuate the functional imaging response to ketamine in healthy subjects. Psychopharmacology (Berl) 2018; 235:1875-1886. [PMID: 29564482 DOI: 10.1007/s00213-018-4877-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 03/08/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND Aberrant glutamate neurotransmission, and in particular dysfunction of the N-methyl-D-aspartate receptor (NMDAR), has been implicated in psychiatric disorders and represents a novel therapeutic target. Low-dose administration of the NMDA antagonist ketamine in healthy volunteers elicits a strong blood oxygenation level dependent (BOLD) imaging signal that can be attenuated by pretreatment with single, therapeutically effective doses of marketed medicines interacting with the glutamate system. OBJECTIVE To test the attenuation of the ketamine-induced BOLD signal by pretreatment with either a metabotropic glutamate receptor (mGluR) 2/3 or a mGluR2 agonist in healthy volunteers METHODS: We used a ketamine challenge pharmacological magnetic resonance imaging (phMRI) paradigm to assess the modulatory effects of single acute doses of LY2140023 (pomaglumetad methionil), the methionine prodrug of the mGluR2/3 agonist LY404039 (10, 40, and 160 mg; N = 16 subjects) and of LY2979165, and the alanine prodrug of the selective orthosteric mGluR2 agonist 2812223 (20 and 60 mg; N = 16 subjects). RESULTS A reduction in the ketamine-evoked BOLD phMRI signal relative to placebo was observed at the highest doses tested of both LY2140023 and LY2979165. A relationship was observed between reduction of the BOLD signal and increasing plasma levels of 2812223 in the LY2979165 cohort. CONCLUSIONS These results identify pharmacologically active doses of the group II mGluR agonist prodrugs LY2140023 and LY2979165 in humans. They also extend the classes of compounds that have been experimentally shown to reverse the ketamine-evoked phMRI signal in humans, further supporting the use of this method as a neuroimaging biomarker for assessing functional effects.
Collapse
Affiliation(s)
- Mitul A Mehta
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK.
| | - Anne Schmechtig
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK
| | - Vasileia Kotoula
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK
| | - Juliet McColm
- Eli Lilly and Company, Sunninghill Road, Windlesham, Surrey, UK
| | | | - Claire Brittain
- Eli Lilly and Company, Sunninghill Road, Windlesham, Surrey, UK
| | | | | | - Paul D Morrison
- Psychosis Studies Department, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Thomas Pollak
- Psychosis Studies Department, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | | | - Steven C R Williams
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK
| | | |
Collapse
|
15
|
Schoenberger M, Schroeder FA, Placzek MS, Carter RL, Rosen BR, Hooker JM, Sander CY. In Vivo [ 18F]GE-179 Brain Signal Does Not Show NMDA-Specific Modulation with Drug Challenges in Rodents and Nonhuman Primates. ACS Chem Neurosci 2018; 9:298-305. [PMID: 29050469 PMCID: PMC5894869 DOI: 10.1021/acschemneuro.7b00327] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
As one of the major excitatory ion channels in the brain, NMDA receptors have been a leading research target for neuroscientists, physicians, medicinal chemists, and pharmaceutical companies for decades. Molecular imaging of NMDA receptors by means of positron emission tomography (PET) with [18F]GE-179 quickly progressed to clinical PET studies, but a thorough understanding of its binding specificity has been missing and has thus limited signal interpretation. Here a preclinical study with [18F]GE-179 in rodents and nonhuman primates (NHPs) is presented in an attempt to characterize [18F]GE-179 signal specificity. Rodent PET/CT was used to study drug occupancy and functional manipulation in rats by pretreating animals with NMDA targeted blocking/modulating drug doses followed by a single bolus of [18F]GE-179. Binding competition with GE-179, MK801, PCP, and ketamine, allosteric inhibition by ifenprodil, and brain activation with methamphetamine did not alter the [18F]GE-179 brain signal in rats. In addition, multimodal imaging with PET/MRI in NHPs was used to evaluate changes in radiotracer binding as a function of pharmacological challenges. Drug-induced hemodynamic changes were monitored simultaneously using functional MRI (fMRI). Comparisons of baseline and signal after drug challenge in NHPs demonstrated that the [18F]GE-179 signal cannot be manipulated in a predictable fashion in vivo. fMRI data acquired simultaneously with PET data supported this finding and provided evidence that radiotracer delivery is not altered by blood flow changes. In conclusion, the [18F]GE-179 brain signal is not readily interpretable in the context of NMDA receptor binding on the basis of the results shown in this study.
Collapse
Affiliation(s)
- Matthias Schoenberger
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital , Charlestown, Massachusetts 02129, United States
- Chemical Biology and Imaging, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven , BE-3000 Leuven, Belgium
| | - Frederick A Schroeder
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital , Charlestown, Massachusetts 02129, United States
| | - Michael S Placzek
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital , Charlestown, Massachusetts 02129, United States
- Department of Psychiatry, McLean Imaging Center, McLean Hospital , Belmont, Massachusetts 02478, United States
- Harvard Medical School , Boston, Massachusetts 02115, United States
| | | | - Bruce R Rosen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital , Charlestown, Massachusetts 02129, United States
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology , Cambridge, Massachusetts 02139, United States
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital , Charlestown, Massachusetts 02129, United States
- Harvard Medical School , Boston, Massachusetts 02115, United States
| | - Christin Y Sander
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital , Charlestown, Massachusetts 02129, United States
- Harvard Medical School , Boston, Massachusetts 02115, United States
| |
Collapse
|
16
|
Tang H, Kukral D, Li YW, Fronheiser M, Malone H, Pena A, Pieschl R, Sidik K, Tobon G, Chow PL, Bristow LJ, Hayes W, Luo F. Mapping the central effects of (±)-ketamine and traxoprodil using pharmacological magnetic resonance imaging in awake rats. J Psychopharmacol 2018; 32:146-155. [PMID: 29378483 DOI: 10.1177/0269881117746901] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Major depressive disorder is a leading cause of disability globally. Improvements in the efficacy of antidepressant therapy are needed as a high proportion (>40%) of individuals with major depressive disorder fail to respond adequately to current treatments. The non-selective N-methyl-D-aspartate receptor channel blocker, (±)-ketamine, has been reported to produce a rapid and long-lasting antidepressant response in treatment-resistant major depressive disorder patients, which provides a unique opportunity for investigation of mechanisms that mediate its therapeutic effect. Efforts have also focused on the development of selective N-methyl-D-aspartate receptor subtype 2B antagonists which may retain antidepressant activity but have lower potential for dissociative/psychotomimetic effects. In the present study, we examined the central nervous system effects of acute, intravenous administration of (±)-ketamine or the N-methyl-D-aspartate receptor subtype 2B antagonist, traxoprodil, in awake rats using pharmacological magnetic resonance imaging. The study contained five treatment groups: vehicle, 3 mg/kg (±)-ketamine, and three doses of traxoprodil (0.3 mg/kg, 5 mg/kg, and 15 mg/kg). Non-linear model fitting was performed on the temporal hemodynamic pharmacological magnetic resonance imaging data to generate brain activation maps as well as regional responses based on blood oxygen level dependent signal changes for group analysis. Traxoprodil at 5 mg/kg and 15 mg/kg produced a dose-dependent pharmacological magnetic resonance imaging signal in rat forebrain regions with both doses achieving >80% N-methyl-D-aspartate receptor subtype 2B occupancy determined by ex vivo [3H]Ro 25-6981 binding. The middle dose of traxoprodil (5 mg/kg) generated region-specific activations in medial prefrontal cortex, ventral orbital cortex, and anterior cingulate cortex whereas the high dose (15 mg/kg) produced a widespread pharmacological magnetic resonance imaging response in both cortical and subcortical brain regions which was similar to that produced by (±)-ketamine (3 mg/kg, intravenous).
Collapse
Affiliation(s)
- Haiying Tang
- 1 Translational Sciences, Bristol-Myers Squibb, Lawrenceville, USA
| | - Daniel Kukral
- 1 Translational Sciences, Bristol-Myers Squibb, Lawrenceville, USA
| | - Yu-Wen Li
- 2 Discovery Biology, Bristol-Myers Squibb, Wallingford, USA
| | | | - Harold Malone
- 1 Translational Sciences, Bristol-Myers Squibb, Lawrenceville, USA
| | - Adrienne Pena
- 1 Translational Sciences, Bristol-Myers Squibb, Lawrenceville, USA
| | - Rick Pieschl
- 2 Discovery Biology, Bristol-Myers Squibb, Wallingford, USA
| | - Kurex Sidik
- 3 Global Biometrics Sciences, Bristol-Myers Squibb, Lawrenceville, USA
| | | | - Patrick L Chow
- 1 Translational Sciences, Bristol-Myers Squibb, Lawrenceville, USA
| | | | - Wendy Hayes
- 1 Translational Sciences, Bristol-Myers Squibb, Lawrenceville, USA
| | - Feng Luo
- 5 Translational Sciences, Bristol-Myers Squibb, Wallingford, USA
| |
Collapse
|
17
|
Carmichael O, Schwarz AJ, Chatham CH, Scott D, Turner JA, Upadhyay J, Coimbra A, Goodman JA, Baumgartner R, English BA, Apolzan JW, Shankapal P, Hawkins KR. The role of fMRI in drug development. Drug Discov Today 2018; 23:333-348. [PMID: 29154758 PMCID: PMC5931333 DOI: 10.1016/j.drudis.2017.11.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/19/2017] [Accepted: 11/13/2017] [Indexed: 12/17/2022]
Abstract
Functional magnetic resonance imaging (fMRI) has been known for over a decade to have the potential to greatly enhance the process of developing novel therapeutic drugs for prevalent health conditions. However, the use of fMRI in drug development continues to be relatively limited because of a variety of technical, biological, and strategic barriers that continue to limit progress. Here, we briefly review the roles that fMRI can have in the drug development process and the requirements it must meet to be useful in this setting. We then provide an update on our current understanding of the strengths and limitations of fMRI as a tool for drug developers and recommend activities to enhance its utility.
Collapse
Affiliation(s)
- Owen Carmichael
- Pennington Biomedical Research Center, Baton Rouge, LA, USA.
| | | | - Christopher H Chatham
- Translational Medicine Neuroscience and Biomarkers, Roche Innovation Center, Basel, Switzerland
| | | | - Jessica A Turner
- Psychology Department & Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | | | | | | | - Richard Baumgartner
- Biostatistics and Research Decision Sciences (BARDS), Merck & Co., Inc., Kenilworth, NJ, USA
| | | | - John W Apolzan
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | | | | |
Collapse
|
18
|
Javitt DC, Carter CS, Krystal JH, Kantrowitz JT, Girgis RR, Kegeles LS, Ragland JD, Maddock RJ, Lesh TA, Tanase C, Corlett PR, Rothman DL, Mason G, Qiu M, Robinson J, Potter WZ, Carlson M, Wall MM, Choo TH, Grinband J, Lieberman JA. Utility of Imaging-Based Biomarkers for Glutamate-Targeted Drug Development in Psychotic Disorders: A Randomized Clinical Trial. JAMA Psychiatry 2018; 75:11-19. [PMID: 29167877 PMCID: PMC5833531 DOI: 10.1001/jamapsychiatry.2017.3572] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
IMPORTANCE Despite strong theoretical rationale and preclinical evidence, several glutamate-targeted treatments for schizophrenia have failed in recent pivotal trials, prompting questions as to target validity, compound inadequacy, or lack of target engagement. A key limitation for glutamate-based treatment development is the lack of functional target-engagement biomarkers for translation between preclinical and early-stage clinical studies. We evaluated the utility of 3 potential biomarkers-ketamine-evoked changes in the functional magnetic imaging (fMRI) blood oxygen level-dependent response (pharmacoBOLD), glutamate proton magnetic resonance spectroscopy (1H MRS), and task-based fMRI-for detecting ketamine-related alterations in brain glutamate. OBJECTIVE To identify measures with sufficient effect size and cross-site reliability to serve as glutamatergic target engagement biomarkers within early-phase clinical studies. DESIGN, SETTING, AND PARTICIPANTS This randomized clinical trial was conducted at an academic research institution between May 2014 and October 2015 as part of the National Institute of Mental Health-funded Fast-Fail Trial for Psychotic Spectrum Disorders project. All raters were blinded to study group. Healthy volunteers aged 18 to 55 years of either sex and free of significant medical or psychiatric history were recruited from 3 sites. Data were analyzed between November 2015 and December 2016. INTERVENTIONS Volunteers received either sequential ketamine (0.23 mg/kg infusion over 1 minute followed by 0.58 mg/kg/h infusion over 30 minutes and then 0.29 mg/kg/h infusion over 29 minutes) or placebo infusions. MAIN OUTCOMES AND MEASURES Ketamine-induced changes in pharmacoBOLD, 1H MRS, and task-based fMRI measures, along with symptom ratings. Measures were prespecified prior to data collection. RESULTS Of the 65 volunteers, 41 (63%) were male, and the mean (SD) age was 31.1 (9.6) years; 59 (91%) had at least 1 valid scan. A total of 53 volunteers (82%) completed both ketamine infusions. In pharmacoBOLD, a highly robust increase (Cohen d = 5.4; P < .001) in fMRI response was observed, with a consistent response across sites. A smaller but significant signal (Cohen d = 0.64; P = .04) was also observed in 1H MRS-determined levels of glutamate+glutamine immediately following ketamine infusion. By contrast, no significant differences in task-activated fMRI responses were found between groups. CONCLUSIONS AND RELEVANCE These findings demonstrate robust effects of ketamine on pharmacoBOLD across sites, supporting its utility for definitive assessment of functional target engagement. Other measures, while sensitive to ketamine effects, were not sufficiently robust for use as cross-site target engagement measures. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT02134951.
Collapse
Affiliation(s)
- Daniel C. Javitt
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York,Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, New York
| | | | - John H. Krystal
- Department of Psychiatry, Yale University, New Haven, Connecticut
| | - Joshua T. Kantrowitz
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York,Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, New York
| | - Ragy R. Girgis
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York
| | - Lawrence S. Kegeles
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York
| | | | | | - Tyler A. Lesh
- Department of Psychiatry, University of California, Davis
| | - Costin Tanase
- Department of Psychiatry, University of California, Davis
| | | | | | - Graeme Mason
- Department of Psychiatry, Yale University, New Haven, Connecticut
| | - Maolin Qiu
- Department of Psychiatry, Yale University, New Haven, Connecticut
| | - James Robinson
- Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, New York
| | | | - Marlene Carlson
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York
| | - Melanie M. Wall
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York,National Institute of Mental Health, Rockville, Maryland
| | - Tse-Hwei Choo
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York
| | - Jack Grinband
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York
| | - Jeffrey A. Lieberman
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York
| |
Collapse
|
19
|
Mei YY, Wu DC, Zhou N. Astrocytic Regulation of Glutamate Transmission in Schizophrenia. Front Psychiatry 2018; 9:544. [PMID: 30459650 PMCID: PMC6232167 DOI: 10.3389/fpsyt.2018.00544] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/12/2018] [Indexed: 01/19/2023] Open
Abstract
According to the glutamate hypothesis of schizophrenia, the abnormality of glutamate transmission induced by hypofunction of NMDA receptors (NMDARs) is causally associated with the positive and negative symptoms of schizophrenia. However, the underlying mechanisms responsible for the changes in glutamate transmission in schizophrenia are not fully understood. Astrocytes, the major regulatory glia in the brain, modulate not only glutamate metabolism but also glutamate transmission. Here we review the recent progress in understanding the role of astrocytes in schizophrenia. We focus on the astrocytic mechanisms of (i) glutamate synthesis via the glutamate-glutamine cycle, (ii) glutamate clearance by excitatory amino acid transporters (EAATs), (iii) D-serine release to activate NMDARs, and (iv) glutamatergic target engagement biomarkers. Abnormality in these processes is highly correlated with schizophrenia phenotypes. These findings will shed light upon further investigation of pathogenesis as well as improvement of biomarkers and therapies for schizophrenia.
Collapse
Affiliation(s)
- Yu-Ying Mei
- Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Dong Chuan Wu
- Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Ning Zhou
- Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
20
|
Paasonen J, Salo RA, Ihalainen J, Leikas JV, Savolainen K, Lehtonen M, Forsberg MM, Gröhn O. Dose-response effect of acute phencyclidine on functional connectivity and dopamine levels, and their association with schizophrenia-like symptom classes in rat. Neuropharmacology 2017; 119:15-25. [DOI: 10.1016/j.neuropharm.2017.03.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/31/2017] [Accepted: 03/20/2017] [Indexed: 10/19/2022]
|
21
|
Kealy J, Commins S, Lowry JP. The effect of NMDA-R antagonism on simultaneously acquired local field potentials and tissue oxygen levels in the brains of freely-moving rats. Neuropharmacology 2017; 116:343-350. [DOI: 10.1016/j.neuropharm.2017.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 12/07/2016] [Accepted: 01/09/2017] [Indexed: 12/14/2022]
|
22
|
Schrantee A, Tremoleda JL, Wylezinska-Arridge M, Bouet V, Hesseling P, Meerhoff GF, de Bruin KM, Koeleman J, Freret T, Boulouard M, Desfosses E, Galineau L, Gozzi A, Dauphin F, Gsell W, Booij J, Lucassen PJ, Reneman L. Repeated dexamphetamine treatment alters the dopaminergic system and increases the phMRI response to methylphenidate. PLoS One 2017; 12:e0172776. [PMID: 28241065 PMCID: PMC5328278 DOI: 10.1371/journal.pone.0172776] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 02/09/2017] [Indexed: 12/12/2022] Open
Abstract
Dexamphetamine (AMPH) is a psychostimulant drug that is used both recreationally and as medication for attention deficit hyperactivity disorder. Preclinical studies have demonstrated that repeated exposure to AMPH can induce damage to nerve terminals of dopamine (DA) neurons. We here assessed the underlying neurobiological changes in the DA system following repeated AMPH exposure and pre-treated rats with AMPH or saline (4 times 5 mg/kg s.c., 2 hours apart), followed by a 1-week washout period. We then used pharmacological MRI (phMRI) with a methylphenidate (MPH) challenge, as a sensitive and non-invasive in-vivo measure of DAergic function. We subsequently validated the DA-ergic changes post-mortem, using a.o. high-performance liquid chromatography (HPLC) and autoradiography. In the AMPH pre-treated group, we observed a significantly larger BOLD response to the MPH challenge, particularly in DA-ergic brain areas and their downstream projections. Subsequent autoradiography studies showed that AMPH pre-treatment significantly reduced DA transporter (DAT) density in the caudate-putamen (CPu) and nucleus accumbens, whereas HPLC analysis revealed increases in the DA metabolite homovanillic acid in the CPu. Our results suggest that AMPH pre-treatment alters DAergic responsivity, a change that can be detected with phMRI in rats. These phMRI changes likely reflect increased DA release together with reduced DAT binding. The ability to assess subtle synaptic changes using phMRI is promising for both preclinical studies of drug discovery, and for clinical studies where phMRI can be a useful tool to non-invasively investigate DA abnormalities, e.g. in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Anouk Schrantee
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Biological Imaging Centre, Imperial College London, White City, London, United Kingdom
- * E-mail:
| | - Jordi L. Tremoleda
- Biological Imaging Centre, Imperial College London, White City, London, United Kingdom
- Centre for Trauma Sciences, The Blizard Institute, London, United Kingdom
| | - Marzena Wylezinska-Arridge
- Biological Imaging Centre, Imperial College London, White City, London, United Kingdom
- Neuroradiological Academic Unit, Department of Brain Repair and Rehabilitation, UCL Institute of Neurology, Queen Square, London, United Kingdom
| | - Valentine Bouet
- Normandie-Université, GMPc, EA 4259, Université de Caen Basse-Normandie, Caen, France
| | - Peter Hesseling
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - Gideon F. Meerhoff
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - Kora M. de Bruin
- Department of Nuclear Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jan Koeleman
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Thomas Freret
- Normandie-Université, GMPc, EA 4259, Université de Caen Basse-Normandie, Caen, France
| | - Michel Boulouard
- Normandie-Université, GMPc, EA 4259, Université de Caen Basse-Normandie, Caen, France
| | - Emilie Desfosses
- UMR Inserm U930, Université François-Rabelais de Tours, Tours, France
| | - Laurent Galineau
- UMR Inserm U930, Université François-Rabelais de Tours, Tours, France
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ UNITN, Rovereto, Italy
| | - François Dauphin
- Normandie-Université, GMPc, EA 4259, Université de Caen Basse-Normandie, Caen, France
| | - Willy Gsell
- Biological Imaging Centre, Imperial College London, White City, London, United Kingdom
- Biomedical MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Jan Booij
- Department of Nuclear Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Paul J. Lucassen
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, The Netherlands
| | - Liesbeth Reneman
- Department of Radiology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Li J, Schwarz AJ, Gilmour G. Relating Translational Neuroimaging and Amperometric Endpoints: Utility for Neuropsychiatric Drug Discovery. Curr Top Behav Neurosci 2016; 28:397-421. [PMID: 27023366 DOI: 10.1007/7854_2016_1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Measures of neuronal activation are a natural and parsimonious translational biomarker to consider in the context of neuropsychiatric drug discovery studies. In this regard, functional neuroimaging using the BOLD fMRI technique is becoming more frequently employed to not only probe aberrant brain regions and circuits in disease, but also to assess the effects of novel pharmacological agents on these processes. In the ideal situation, these types of studies would first be conducted pre-clinically in rodents to confirm a measurable functional response on relevant brain circuits before seeking to replicate the findings in an analogous fMRI paradigm in humans. However, the need for animal immobilization during the scanning procedure precludes all but the simplest behavioural task-based paradigms in rodent BOLD fMRI. This chapter considers how in vivo oxygen amperometry may represent a viable and valid proxy for BOLD fMRI in freely moving rodents engaged in behavioural tasks. The amperometric technique and several examples of emerging evidence are described to show how the technique can deliver results that translate to pharmacological, event-related and functional connectivity variants of fMRI. In vivo oxygen amperometry holds great promise as a technique that may help to bridge the gap between basic drug discovery research in rodents and applied efficacy testing in humans.
Collapse
Affiliation(s)
- Jennifer Li
- In Vivo Pharmacology, Eli Lilly and Company, Erl Wood Manor, Sunninghill Road, Windlesham, UK
| | - Adam J Schwarz
- Translational Imaging, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Gary Gilmour
- In Vivo Pharmacology, Eli Lilly and Company, Erl Wood Manor, Sunninghill Road, Windlesham, UK.
| |
Collapse
|
24
|
Shcherbinin S, Doyle O, Zelaya FO, de Simoni S, Mehta MA, Schwarz AJ. Modulatory effects of ketamine, risperidone and lamotrigine on resting brain perfusion in healthy human subjects. Psychopharmacology (Berl) 2015. [PMID: 26223493 DOI: 10.1007/s00213-015-4021-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
RATIONALE Resting brain perfusion, measured using the MRI-based arterial spin labelling (ASL) technique, is sensitive to detect central effects of single, clinically effective, doses of pharmacological compounds. However, pharmacological interaction experiments, such as the modulation of one drug response in the presence of another, have not been widely investigated using a task-free ASL approach. OBJECTIVES We assessed the effects of three psychoactive compounds (ketamine, risperidone and lamotrigine), and their interaction, on resting brain perfusion in healthy human volunteers. METHODS A multivariate Gaussian process classification (GPC) and more conventional univariate analyses were applied. The four pre-infusion conditions for each subject comprised risperidone, lamotrigine and two placebo sessions. The two placebo conditions enabled us to evaluate the classification performance in a test-retest setting, in addition to its performance in distinguishing the active oral drugs from placebo (direct effect on brain perfusion). The post ketamine- or saline-infusion scans allowed the effect of ketamine, and its interaction with risperidone and lamotrigine, on brain perfusion to be characterised. RESULTS The pseudo-continuous ASL measurements of perfusion were sensitive to the effects of ketamine infusion and risperidone. The GPC captured consistent changes in perfusion across the group and contextualised the univariate changes with a larger pattern of regions contributing to accurate discrimination of ketamine from placebo. CONCLUSIONS The findings argue against perfusion changes confounding in the previously described evoked BOLD response to ketamine and emphasise the blockade of the NMDA receptor over neuronal glutamate release in determining the perfusion changes induced by ketamine.
Collapse
Affiliation(s)
- Sergey Shcherbinin
- Tailored Therapeutics - Neuroscience, Eli Lilly and Company, Indianapolis, IN, 46285, USA
| | - Orla Doyle
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Fernando O Zelaya
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Sara de Simoni
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Mitul A Mehta
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, UK
| | - Adam J Schwarz
- Tailored Therapeutics - Neuroscience, Eli Lilly and Company, Indianapolis, IN, 46285, USA.
| |
Collapse
|
25
|
Francois J, Gastambide F, Conway MW, Tricklebank M, Gilmour G. Dissociation of mGlu2/3 agonist effects on ketamine-induced regional and event-related oxygen signals. Psychopharmacology (Berl) 2015; 232:4219-29. [PMID: 25943169 DOI: 10.1007/s00213-015-3948-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 04/20/2015] [Indexed: 12/22/2022]
Abstract
RATIONALE Validating preclinical biomarkers that predict treatment efficacy remains a critical imperative for neuropsychiatric drug discovery. With the establishment of novel in vivo imaging methods, it has become possible to think how such translational proof-of-concept studies may look. OBJECTIVES The aim of this study was to use in vivo oxygen (O2) amperometry to simultaneously assess the regional and event/task-related O2 changes induced by ketamine challenge in rats, and to determine whether both of these signals are equivalently affected by the mGlu2/3 receptor agonist LY379268. METHODS O2 signals were measured via carbon paste electrodes implanted in the anterior cingulate cortex (ACC) of rats trained to perform a simple reaction time task (SRT). SRT performance, event-related ACC O2 responses, and regional ACC O2 signal were recorded simultaneously in animals treated with ketamine (10 mg/kg) and/or LY379268 (3 mg/kg). RESULTS A consistent relationship was observed between baseline SRT performance and related ACC O2 signals, suggesting that ACC engagement is likely to be a requirement for optimal task performance. Ketamine induced a robust and consistent slowing in reaction times that was reflected by a delayed event-related ACC O2 signal increase compared to vehicle controls. Ketamine also produced a regional and task-independent 60-min increase in ACC O2 levels which was effectively attenuated by LY379268. However, LY379238 failed to reverse alterations in event-related O2 signals and associated SRT task performance. CONCLUSIONS These findings raise questions about the degree to which such reversals of regional ketamine O2 signals could potentially be claimed to predict drug treatment efficacy.
Collapse
Affiliation(s)
- Jennifer Francois
- In Vivo Pharmacology, Lilly Research Laboratories, Eli Lilly & Co. Ltd, Erl Wood Manor, Sunninghill Road, Windlesham, Surrey, GU20 6PH, England, UK
| | - Francois Gastambide
- In Vivo Pharmacology, Lilly Research Laboratories, Eli Lilly & Co. Ltd, Erl Wood Manor, Sunninghill Road, Windlesham, Surrey, GU20 6PH, England, UK.
| | - Michael Warwick Conway
- In Vivo Pharmacology, Lilly Research Laboratories, Eli Lilly & Co. Ltd, Erl Wood Manor, Sunninghill Road, Windlesham, Surrey, GU20 6PH, England, UK
| | - Mark Tricklebank
- In Vivo Pharmacology, Lilly Research Laboratories, Eli Lilly & Co. Ltd, Erl Wood Manor, Sunninghill Road, Windlesham, Surrey, GU20 6PH, England, UK
| | - Gary Gilmour
- In Vivo Pharmacology, Lilly Research Laboratories, Eli Lilly & Co. Ltd, Erl Wood Manor, Sunninghill Road, Windlesham, Surrey, GU20 6PH, England, UK
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW The ketamine model has dominated drug discovery in schizophrenia over the past decade, supported by genetic and postmortem evidence implicating glutamatergic transmission. This review assesses recent successes and disappointments of glutamatergic agents and identifies promising new directions. RECENT FINDINGS Strategies focused on enhancing activity of the N-methyl D-aspartate (NMDA) receptor via direct agonists at the glycine site or by inhibition of glycine reuptake have produced modest and often inconsistent evidence of efficacy, as have approaches to reduce excessive glutamate release by lamotrigine or by mGluR2/3 agonists. Strategies targeting α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors have also met with only limited success. Newer approaches include selective allosteric modulation of NMDA receptor subunits and of mGluR5 receptors. In addition, intracellular pathways downstream of NMDA receptors may also provide new treatment targets, as exemplified by phosphodiesterase (PDE) inhibitors. SUMMARY Targeting glutamatergic transmission remains one of the most promising strategies in schizophrenia, particularly early in the course of illness, but therapeutic approaches may require greater specificity for receptor subtype type, illness phase, and individual biology in order to enhance efficacy and overcome problems with reproducibility of clinical results.
Collapse
|
27
|
Hegedűs N, Laszy J, Gyertyán I, Kocsis P, Gajári D, Dávid S, Deli L, Pozsgay Z, Tihanyi K. Scopolamine provocation-based pharmacological MRI model for testing procognitive agents. J Psychopharmacol 2015; 29:447-55. [PMID: 25586394 DOI: 10.1177/0269881114565652] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
There is a huge unmet need to understand and treat pathological cognitive impairment. The development of disease modifying cognitive enhancers is hindered by the lack of correct pathomechanism and suitable animal models. Most animal models to study cognition and pathology do not fulfil either the predictive validity, face validity or construct validity criteria, and also outcome measures greatly differ from those of human trials. Fortunately, some pharmacological agents such as scopolamine evoke similar effects on cognition and cerebral circulation in rodents and humans and functional MRI enables us to compare cognitive agents directly in different species. In this paper we report the validation of a scopolamine based rodent pharmacological MRI provocation model. The effects of deemed procognitive agents (donepezil, vinpocetine, piracetam, alpha 7 selective cholinergic compounds EVP-6124, PNU-120596) were compared on the blood-oxygen-level dependent responses and also linked to rodent cognitive models. These drugs revealed significant effect on scopolamine induced blood-oxygen-level dependent change except for piracetam. In the water labyrinth test only PNU-120596 did not show a significant effect. This provocational model is suitable for testing procognitive compounds. These functional MR imaging experiments can be paralleled with human studies, which may help reduce the number of false cognitive clinical trials.
Collapse
Affiliation(s)
- Nikolett Hegedűs
- Preclinical Imaging Centre, Gedeon Richter Plc, Budapest, Hungary
| | - Judit Laszy
- Department of Behavioural Pharmacology, Gedeon Richter Plc, Budapest, Hungary
| | - István Gyertyán
- Department of Behavioural Pharmacology, Gedeon Richter Plc, Budapest, Hungary
| | - Pál Kocsis
- Preclinical Imaging Centre, Gedeon Richter Plc, Budapest, Hungary
| | - Dávid Gajári
- Preclinical Imaging Centre, Gedeon Richter Plc, Budapest, Hungary
| | - Szabolcs Dávid
- Preclinical Imaging Centre, Gedeon Richter Plc, Budapest, Hungary
| | - Levente Deli
- Preclinical Imaging Centre, Gedeon Richter Plc, Budapest, Hungary
| | - Zsófia Pozsgay
- Preclinical Imaging Centre, Gedeon Richter Plc, Budapest, Hungary
| | - Károly Tihanyi
- Preclinical Imaging Centre, Gedeon Richter Plc, Budapest, Hungary
| |
Collapse
|
28
|
Errico F, D'Argenio V, Sforazzini F, Iasevoli F, Squillace M, Guerri G, Napolitano F, Angrisano T, Di Maio A, Keller S, Vitucci D, Galbusera A, Chiariotti L, Bertolino A, de Bartolomeis A, Salvatore F, Gozzi A, Usiello A. A role for D-aspartate oxidase in schizophrenia and in schizophrenia-related symptoms induced by phencyclidine in mice. Transl Psychiatry 2015; 5:e512. [PMID: 25689573 PMCID: PMC4445752 DOI: 10.1038/tp.2015.2] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 12/19/2014] [Accepted: 12/19/2014] [Indexed: 01/19/2023] Open
Abstract
Increasing evidence points to a role for dysfunctional glutamate N-methyl-D-aspartate receptor (NMDAR) neurotransmission in schizophrenia. D-aspartate is an atypical amino acid that activates NMDARs through binding to the glutamate site on GluN2 subunits. D-aspartate is present in high amounts in the embryonic brain of mammals and rapidly decreases after birth, due to the activity of the enzyme D-aspartate oxidase (DDO). The agonistic activity exerted by D-aspartate on NMDARs and its neurodevelopmental occurrence make this D-amino acid a potential mediator for some of the NMDAR-related alterations observed in schizophrenia. Consistently, substantial reductions of D-aspartate and NMDA were recently observed in the postmortem prefrontal cortex of schizophrenic patients. Here we show that DDO mRNA expression is increased in prefrontal samples of schizophrenic patients, thus suggesting a plausible molecular event responsible for the D-aspartate imbalance previously described. To investigate whether altered D-aspartate levels can modulate schizophrenia-relevant circuits and behaviors, we also measured the psychotomimetic effects produced by the NMDAR antagonist, phencyclidine, in Ddo knockout mice (Ddo(-)(/-)), an animal model characterized by tonically increased D-aspartate levels since perinatal life. We show that Ddo(-/-) mice display a significant reduction in motor hyperactivity and prepulse inhibition deficit induced by phencyclidine, compared with controls. Furthermore, we reveal that increased levels of D-aspartate in Ddo(-/-) animals can significantly inhibit functional circuits activated by phencyclidine, and affect the development of cortico-hippocampal connectivity networks potentially involved in schizophrenia. Collectively, the present results suggest that altered D-aspartate levels can influence neurodevelopmental brain processes relevant to schizophrenia.
Collapse
Affiliation(s)
- F Errico
- Ceinge Biotecnologie Avanzate, Naples, Italy,Department of Molecular Medicine and Medical Biotechnology, University of Naples ‘Federico II', Naples, Italy,Ceinge Biotecnologie Avanzate, Via G. Salvatore, 486, 80145 Naples, Italy E-mail:
| | - V D'Argenio
- Ceinge Biotecnologie Avanzate, Naples, Italy,Department of Molecular Medicine and Medical Biotechnology, University of Naples ‘Federico II', Naples, Italy
| | - F Sforazzini
- Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto, Italy
| | - F Iasevoli
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine ‘Federico II', Naples, Italy
| | - M Squillace
- Ceinge Biotecnologie Avanzate, Naples, Italy
| | - G Guerri
- Ceinge Biotecnologie Avanzate, Naples, Italy
| | - F Napolitano
- Ceinge Biotecnologie Avanzate, Naples, Italy,Department of Molecular Medicine and Medical Biotechnology, University of Naples ‘Federico II', Naples, Italy
| | - T Angrisano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples ‘Federico II', Naples, Italy,IEOS, CNR, Naples, Italy,Department of Biology, University of Naples ‘Federico II', Naples, Italy
| | - A Di Maio
- Ceinge Biotecnologie Avanzate, Naples, Italy
| | - S Keller
- Department of Molecular Medicine and Medical Biotechnology, University of Naples ‘Federico II', Naples, Italy,IEOS, CNR, Naples, Italy
| | - D Vitucci
- Ceinge Biotecnologie Avanzate, Naples, Italy
| | - A Galbusera
- Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto, Italy
| | - L Chiariotti
- Department of Molecular Medicine and Medical Biotechnology, University of Naples ‘Federico II', Naples, Italy,IEOS, CNR, Naples, Italy
| | - A Bertolino
- Group of Psychiatric Neuroscience, Department of Neuroscience, Basic Sciences and Sense Organs, University of Bari ‘Aldo Moro', Bari, Italy,pRED, Neuroscience DTA, Hoffman-La Roche, Ltd, Basel, Switzerland
| | - A de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry, Department of Neuroscience, University School of Medicine ‘Federico II', Naples, Italy
| | - F Salvatore
- Ceinge Biotecnologie Avanzate, Naples, Italy,Department of Molecular Medicine and Medical Biotechnology, University of Naples ‘Federico II', Naples, Italy,IRCCS-Fondazione SDN, Via Gianturco, Naples, Italy
| | - A Gozzi
- Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto, Italy,Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Corso Bettini, 31, 38068 Rovereto, Italy. E-mail:
| | - A Usiello
- Ceinge Biotecnologie Avanzate, Naples, Italy,Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Second University of Naples (SUN), Caserta, Italy
| |
Collapse
|
29
|
Barnes SA, Sawiak SJ, Caprioli D, Jupp B, Buonincontri G, Mar AC, Harte MK, Fletcher PC, Robbins TW, Neill JC, Dalley JW. Impaired limbic cortico-striatal structure and sustained visual attention in a rodent model of schizophrenia. Int J Neuropsychopharmacol 2014; 18:pyu010. [PMID: 25552430 PMCID: PMC4368881 DOI: 10.1093/ijnp/pyu010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 06/09/2014] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND N-methyl-d-aspartate receptor (NMDAR) dysfunction is thought to contribute to the pathophysiology of schizophrenia. Accordingly, NMDAR antagonists such as phencyclidine (PCP) are used widely in experimental animals to model cognitive impairment associated with this disorder. However, it is unclear whether PCP disrupts the structural integrity of brain areas relevant to the profile of cognitive impairment in schizophrenia. METHODS Here we used high-resolution magnetic resonance imaging and voxel-based morphometry to investigate structural alterations associated with sub-chronic PCP treatment in rats. RESULTS Sub-chronic exposure of rats to PCP (5mg/kg twice daily for 7 days) impaired sustained visual attention on a 5-choice serial reaction time task, notably when the attentional load was increased. In contrast, sub-chronic PCP had no significant effect on the attentional filtering of a pre-pulse auditory stimulus in an acoustic startle paradigm. Voxel-based morphometry revealed significantly reduced grey matter density bilaterally in the hippocampus, anterior cingulate cortex, ventral striatum, and amygdala. PCP-treated rats also exhibited reduced cortical thickness in the insular cortex. CONCLUSIONS These findings demonstrate that sub-chronic NMDA receptor antagonism is sufficient to produce highly-localized morphological abnormalities in brain areas implicated in the pathogenesis of schizophrenia. Furthermore, PCP exposure resulted in dissociable impairments in attentional function.
Collapse
Affiliation(s)
- Samuel A Barnes
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA (Dr Barnes); Behavioural and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Downing St, Cambridge UK (Drs Sawiak, Caprioli, Jupp, Mar, Fletcher, Robbins, and Dalley); Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Sawiak and Buonincontri); Department of Psychiatry, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Fletcher and Dalley); Manchester Pharmacy School, University of Manchester, UK (Drs Harte and Neill)
| | - Stephen J Sawiak
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA (Dr Barnes); Behavioural and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Downing St, Cambridge UK (Drs Sawiak, Caprioli, Jupp, Mar, Fletcher, Robbins, and Dalley); Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Sawiak and Buonincontri); Department of Psychiatry, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Fletcher and Dalley); Manchester Pharmacy School, University of Manchester, UK (Drs Harte and Neill)
| | - Daniele Caprioli
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA (Dr Barnes); Behavioural and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Downing St, Cambridge UK (Drs Sawiak, Caprioli, Jupp, Mar, Fletcher, Robbins, and Dalley); Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Sawiak and Buonincontri); Department of Psychiatry, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Fletcher and Dalley); Manchester Pharmacy School, University of Manchester, UK (Drs Harte and Neill)
| | - Bianca Jupp
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA (Dr Barnes); Behavioural and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Downing St, Cambridge UK (Drs Sawiak, Caprioli, Jupp, Mar, Fletcher, Robbins, and Dalley); Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Sawiak and Buonincontri); Department of Psychiatry, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Fletcher and Dalley); Manchester Pharmacy School, University of Manchester, UK (Drs Harte and Neill)
| | - Guido Buonincontri
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA (Dr Barnes); Behavioural and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Downing St, Cambridge UK (Drs Sawiak, Caprioli, Jupp, Mar, Fletcher, Robbins, and Dalley); Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Sawiak and Buonincontri); Department of Psychiatry, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Fletcher and Dalley); Manchester Pharmacy School, University of Manchester, UK (Drs Harte and Neill)
| | - Adam C Mar
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA (Dr Barnes); Behavioural and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Downing St, Cambridge UK (Drs Sawiak, Caprioli, Jupp, Mar, Fletcher, Robbins, and Dalley); Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Sawiak and Buonincontri); Department of Psychiatry, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Fletcher and Dalley); Manchester Pharmacy School, University of Manchester, UK (Drs Harte and Neill)
| | - Michael K Harte
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA (Dr Barnes); Behavioural and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Downing St, Cambridge UK (Drs Sawiak, Caprioli, Jupp, Mar, Fletcher, Robbins, and Dalley); Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Sawiak and Buonincontri); Department of Psychiatry, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Fletcher and Dalley); Manchester Pharmacy School, University of Manchester, UK (Drs Harte and Neill)
| | - Paul C Fletcher
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA (Dr Barnes); Behavioural and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Downing St, Cambridge UK (Drs Sawiak, Caprioli, Jupp, Mar, Fletcher, Robbins, and Dalley); Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Sawiak and Buonincontri); Department of Psychiatry, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Fletcher and Dalley); Manchester Pharmacy School, University of Manchester, UK (Drs Harte and Neill)
| | - Trevor W Robbins
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA (Dr Barnes); Behavioural and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Downing St, Cambridge UK (Drs Sawiak, Caprioli, Jupp, Mar, Fletcher, Robbins, and Dalley); Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Sawiak and Buonincontri); Department of Psychiatry, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Fletcher and Dalley); Manchester Pharmacy School, University of Manchester, UK (Drs Harte and Neill)
| | - Jo C Neill
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA (Dr Barnes); Behavioural and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Downing St, Cambridge UK (Drs Sawiak, Caprioli, Jupp, Mar, Fletcher, Robbins, and Dalley); Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Sawiak and Buonincontri); Department of Psychiatry, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Fletcher and Dalley); Manchester Pharmacy School, University of Manchester, UK (Drs Harte and Neill)
| | - Jeffrey W Dalley
- Department of Psychiatry, School of Medicine, University of California San Diego, La Jolla, CA (Dr Barnes); Behavioural and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Downing St, Cambridge UK (Drs Sawiak, Caprioli, Jupp, Mar, Fletcher, Robbins, and Dalley); Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Sawiak and Buonincontri); Department of Psychiatry, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK (Drs Fletcher and Dalley); Manchester Pharmacy School, University of Manchester, UK (Drs Harte and Neill).
| |
Collapse
|
30
|
Hiyoshi T, Marumo T, Hikichi H, Tomishima Y, Urabe H, Tamita T, Iida I, Yasuhara A, Karasawa JI, Chaki S. Neurophysiologic and antipsychotic profiles of TASP0433864, a novel positive allosteric modulator of metabotropic glutamate 2 receptor. J Pharmacol Exp Ther 2014; 351:642-53. [PMID: 25277141 DOI: 10.1124/jpet.114.218651] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Excess glutamatergic neurotransmission has been implicated in the pathophysiology of schizophrenia, and the activation of metabotropic glutamate 2 (mGlu2) receptor may exert antipsychotic effects by normalizing glutamate transmission. In the present study, we investigated the neurophysiologic and antipsychotic profiles of TASP0433864 [(2S)-2-[(4-tert-butylphenoxy)methyl]-5-methyl-2,3-dihydroimidazo[2,1-b][1,3]oxazole-6-carboxamide], a newly synthesized positive allosteric modulator (PAM) of mGlu2 receptor. TASP0433864 exhibited PAM activity at human and rat mGlu2 receptors with EC50 values of 199 and 206 nM, respectively, without exerting agonist activity at rat mGlu2 receptor. TASP0433864 produced a leftward and upward shift in the concentration-response curve of glutamate-increased guanosine 5'-O-(3-[(35)S]thio)triphosphate binding to mGlu2 receptor. In contrast, TASP0433864 had negligible activities for other mGlu receptors, including mGlu3 receptor, and did not have any affinity for other receptors or transporters. In hippocampal slices, TASP0433864 potentiated an inhibitory effect of DCG-IV [(2S,2'R,3'R)-2-(2',3'-dicarboxylcyclopropyl)glycine], a mGlu2/3 receptor agonist, on the field excitatory postsynaptic potentials in the dentate gyrus, indicating that TASP0433864 potentiates the mGlu2 receptor-mediated presynaptic inhibition of glutamate release. Moreover, TASP0433864 inhibited both MK-801 [(5S,10R)-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine hydrogen maleate]- and ketamine-increased cortical γ band oscillation in the rat cortical electroencephalogram, which have been considered to reflect the excess activation of cortical pyramidal neurons. The inhibitory effect of TASP0433864 on cortical activation was also observed in the mouse 2-deoxy-glucose uptake study. In a behavioral study, TASP0433864 significantly inhibited both ketamine- and methamphetamine-increased locomotor activities in mice and rats, respectively. Collectively, these findings indicate that TASP0433864 is a selective mGlu2 receptor PAM with antipsychotic activity, and the attenuation of excess glutamatergic neurotransmission may be involved in the action of TASP0433864.
Collapse
Affiliation(s)
- Tetsuaki Hiyoshi
- Pharmacology Laboratories (T.H., T.M., H.H., Y.T., J.K., S.C.), Chemistry Laboratories (H.U., T.T., A.Y.), and Drug Safety and Pharmacokinetics Laboratories (I.I.), Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Toshiyuki Marumo
- Pharmacology Laboratories (T.H., T.M., H.H., Y.T., J.K., S.C.), Chemistry Laboratories (H.U., T.T., A.Y.), and Drug Safety and Pharmacokinetics Laboratories (I.I.), Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Hirohiko Hikichi
- Pharmacology Laboratories (T.H., T.M., H.H., Y.T., J.K., S.C.), Chemistry Laboratories (H.U., T.T., A.Y.), and Drug Safety and Pharmacokinetics Laboratories (I.I.), Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Yasumitsu Tomishima
- Pharmacology Laboratories (T.H., T.M., H.H., Y.T., J.K., S.C.), Chemistry Laboratories (H.U., T.T., A.Y.), and Drug Safety and Pharmacokinetics Laboratories (I.I.), Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Hiroki Urabe
- Pharmacology Laboratories (T.H., T.M., H.H., Y.T., J.K., S.C.), Chemistry Laboratories (H.U., T.T., A.Y.), and Drug Safety and Pharmacokinetics Laboratories (I.I.), Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Tomoko Tamita
- Pharmacology Laboratories (T.H., T.M., H.H., Y.T., J.K., S.C.), Chemistry Laboratories (H.U., T.T., A.Y.), and Drug Safety and Pharmacokinetics Laboratories (I.I.), Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Izumi Iida
- Pharmacology Laboratories (T.H., T.M., H.H., Y.T., J.K., S.C.), Chemistry Laboratories (H.U., T.T., A.Y.), and Drug Safety and Pharmacokinetics Laboratories (I.I.), Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Akito Yasuhara
- Pharmacology Laboratories (T.H., T.M., H.H., Y.T., J.K., S.C.), Chemistry Laboratories (H.U., T.T., A.Y.), and Drug Safety and Pharmacokinetics Laboratories (I.I.), Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Jun-ichi Karasawa
- Pharmacology Laboratories (T.H., T.M., H.H., Y.T., J.K., S.C.), Chemistry Laboratories (H.U., T.T., A.Y.), and Drug Safety and Pharmacokinetics Laboratories (I.I.), Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Shigeyuki Chaki
- Pharmacology Laboratories (T.H., T.M., H.H., Y.T., J.K., S.C.), Chemistry Laboratories (H.U., T.T., A.Y.), and Drug Safety and Pharmacokinetics Laboratories (I.I.), Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| |
Collapse
|
31
|
Hiyoshi T, Kambe D, Karasawa J, Chaki S. Involvement of glutamatergic and GABAergic transmission in MK-801-increased gamma band oscillation power in rat cortical electroencephalograms. Neuroscience 2014; 280:262-74. [PMID: 25220900 DOI: 10.1016/j.neuroscience.2014.08.047] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 08/25/2014] [Accepted: 08/31/2014] [Indexed: 12/30/2022]
Abstract
Hypofunction of the N-methyl-D-aspartic acid receptor (NMDAr) has been considered to play a crucial role in the pathophysiology of schizophrenia. In rodent electroencephalogram (EEG) studies, non-competitive NMDAr antagonists have been reported to produce aberrant basal gamma band oscillation (GBO), as observed in schizophrenia. Aberrations in GBO power have attracted attention as a translational biomarker for the development of novel antipsychotic drugs. However, the neuronal mechanisms as well as the pharmacological significance of NMDAr antagonist-induced aberrant GBO power have not been fully investigated. In the present study, to address the above questions, we examined the pharmacological properties of MK-801 (0.1 mg/kg)-increased basal GBO power in rat cortical EEG. Riluzole (3-10 mg/kg), a glutamate release inhibitor, reduced the MK-801-increased basal GBO power. In contrast, L-838,417 (1-3 mg/kg), an α2/3/5 subunit-selective GABAA receptor-positive allosteric modulator, enhanced the GBO increase. Antipsychotics such as haloperidol (0.05-0.3 mg/kg) and clozapine (1-10 mg/kg) dose-dependently attenuated the MK-801-increased GBO power. Likewise, LY379268 (0.3-3 mg/kg), an metabotropic glutamate 2/3 receptor (mGlu2/3 receptor) agonist, reduced the GBO increase in a dose-dependent manner, which was antagonized by an mGlu2/3 receptor antagonist LY341495. These results suggest that an increase in cortical GBO power induced by NMDAr hypofunction can be attributed to the aberrant activities of both excitatory pyramidal neurons and inhibitory interneurons in local circuits. The aberrant cortical GBO power reflecting cortical network dysfunction observed in schizophrenia might be a useful biomarker for the discovery of novel antipsychotic drugs.
Collapse
Affiliation(s)
- T Hiyoshi
- Pharmacology 1, Pharmacology Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama, Saitama 331-9530, Japan.
| | - D Kambe
- Pharmacology 1, Pharmacology Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama, Saitama 331-9530, Japan
| | - J Karasawa
- Pharmacology 1, Pharmacology Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama, Saitama 331-9530, Japan
| | - S Chaki
- Pharmacology 1, Pharmacology Laboratories, Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku, Saitama, Saitama 331-9530, Japan
| |
Collapse
|
32
|
Li J, Ishiwari K, Conway MW, Francois J, Huxter J, Lowry JP, Schwarz AJ, Tricklebank M, Gilmour G. Dissociable effects of antipsychotics on ketamine-induced changes in regional oxygenation and inter-regional coherence of low frequency oxygen fluctuations in the rat. Neuropsychopharmacology 2014; 39:1635-44. [PMID: 24442094 PMCID: PMC4023136 DOI: 10.1038/npp.2014.10] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 01/13/2014] [Accepted: 01/14/2014] [Indexed: 11/08/2022]
Abstract
Typical and atypical antipsychotics have been shown to alleviate N-methyl-D-aspartate (NMDA) receptor antagonist-induced BOLD signals in healthy humans and animals to differing degrees; factors that might relate to their different molecular mechanisms and clinical profiles. Recent studies have also extended these investigations to the analysis of resting state functional connectivity measures of BOLD signals in different brain regions. Using constant potential amperometry, we examined the effects of the NMDA receptor antagonist S-(+)-ketamine on tissue oxygen levels in medial prefrontal cortex (mPFC) and medial ventral striatum (mVS), and temporal coherence of low-frequency oxygen fluctuations between these regions in freely moving rats. Furthermore, we assessed the extent to which the atypical antipsychotic clozapine and the typical antipsychotic haloperidol could modulate the effects of S-(+)-ketamine on these measures. Acute S-(+)-ketamine (5-25 mg/kg) produced dose-dependent increases in both tissue O2 levels and coherence. Although effects of clozapine and haloperidol alone were relatively minor, their effects on ketamine-induced signals were markedly more distinct. Clozapine dose-dependently attenuated the absolute S-(+)-ketamine (25 mg/kg) O2 signal in both regions, and also attenuated ketamine-induced increases in regional coherence. Haloperidol had no effect on the absolute ketamine O2 signal yet potentiated increases in regional coherence. The dissociable effects of haloperidol and clozapine on ketamine-induced hyperoxygenation and mPFC-mVS coherence elucidate potentially important mechanistic differences between these classes of pharmacology. This study demonstrates for the first time that in vivo amperometry can measure both regional brain tissue O2 levels and inter-regional coherence, advancing BOLD-like measurements of functional connectivity into awake, unconstrained animals.
Collapse
Affiliation(s)
- Jennifer Li
- Centre for Cognitive Neuroscience, Eli Lilly and Co. Limited, Windlesham, Surrey, UK
| | - Keita Ishiwari
- Centre for Cognitive Neuroscience, Eli Lilly and Co. Limited, Windlesham, Surrey, UK
- Department of Chemistry, National University of Ireland, Maynooth, Co. Kildare, Ireland
| | - Michael W Conway
- Centre for Cognitive Neuroscience, Eli Lilly and Co. Limited, Windlesham, Surrey, UK
| | - Jennifer Francois
- Centre for Cognitive Neuroscience, Eli Lilly and Co. Limited, Windlesham, Surrey, UK
| | - John Huxter
- Centre for Cognitive Neuroscience, Eli Lilly and Co. Limited, Windlesham, Surrey, UK
| | - John P Lowry
- Department of Chemistry, National University of Ireland, Maynooth, Co. Kildare, Ireland
| | - Adam J Schwarz
- Tailored Therapeutics, Eli Lilly and Company, Indianapolis, IN, USA
| | - Mark Tricklebank
- Centre for Cognitive Neuroscience, Eli Lilly and Co. Limited, Windlesham, Surrey, UK
| | - Gary Gilmour
- Centre for Cognitive Neuroscience, Eli Lilly and Co. Limited, Windlesham, Surrey, UK
| |
Collapse
|
33
|
Broberg BV, Madsen KH, Plath N, Olsen CK, Glenthøj BY, Paulson OB, Bjelke B, Søgaard LV. A schizophrenia rat model induced by early postnatal phencyclidine treatment and characterized by Magnetic Resonance Imaging. Behav Brain Res 2013; 250:1-8. [DOI: 10.1016/j.bbr.2013.04.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 04/18/2013] [Accepted: 04/20/2013] [Indexed: 12/15/2022]
|
34
|
Hooker B, Tobon G, Baker S, Zhu C, Hesterman J, Schmidt K, Rajagovindan R, Chandran P, Joshi S, Bannon A, Hoppin J, Beaver J, Fox G, Day M, Upadhyay J. Gabapentin-induced pharmacodynamic effects in the spinal nerve ligation model of neuropathic pain. Eur J Pain 2013; 18:223-37. [DOI: 10.1002/j.1532-2149.2013.00364.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2013] [Indexed: 12/15/2022]
Affiliation(s)
- B.A. Hooker
- Integrated Science and Technology; Abbvie Inc.; North Chicago USA
| | | | - S.J. Baker
- Integrated Science and Technology; Abbvie Inc.; North Chicago USA
| | - C. Zhu
- Neuroscience Discovery; Abbvie Inc.; North Chicago USA
| | | | | | - R. Rajagovindan
- Integrated Science and Technology; Abbvie Inc.; North Chicago USA
| | - P. Chandran
- Integrated Science and Technology; Abbvie Inc.; North Chicago USA
| | - S.K. Joshi
- Neuroscience Discovery; Abbvie Inc.; North Chicago USA
| | - A.W. Bannon
- Neuroscience Discovery; Abbvie Inc.; North Chicago USA
| | | | - J. Beaver
- Integrated Science and Technology; Abbvie Inc.; North Chicago USA
| | - G.B. Fox
- Integrated Science and Technology; Abbvie Inc.; North Chicago USA
| | - M. Day
- Integrated Science and Technology; Abbvie Inc.; North Chicago USA
| | - J. Upadhyay
- Integrated Science and Technology; Abbvie Inc.; North Chicago USA
| |
Collapse
|
35
|
Imaging patients with psychosis and a mouse model establishes a spreading pattern of hippocampal dysfunction and implicates glutamate as a driver. Neuron 2013; 78:81-93. [PMID: 23583108 DOI: 10.1016/j.neuron.2013.02.011] [Citation(s) in RCA: 394] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2013] [Indexed: 11/22/2022]
Abstract
VIDEO ABSTRACT The hippocampus in schizophrenia is characterized by both hypermetabolism and reduced size. It remains unknown whether these abnormalities are mechanistically linked. Here we addressed this question by using MRI tools that can map hippocampal metabolism and structure in patients and mouse models. In at-risk patients, hypermetabolism was found to begin in CA1 and spread to the subiculum after psychosis onset. CA1 hypermetabolism at baseline predicted hippocampal atrophy, which occurred during progression to psychosis, most prominently in similar regions. Next, we used ketamine to model conditions of acute psychosis in mice. Acute ketamine reproduced a similar regional pattern of hypermetabolism, while repeated exposure shifted the hippocampus to a hypermetabolic basal state with concurrent atrophy and pathology in parvalbumin-expressing interneurons. Parallel in vivo experiments using the glutamate-reducing drug LY379268 and direct measurements of extracellular glutamate showed that glutamate drives both neuroimaging abnormalities. These findings show that hippocampal hypermetabolism leads to atrophy in psychotic disorder and suggest glutamate as a pathogenic driver.
Collapse
|
36
|
Sekar S, Jonckers E, Verhoye M, Willems R, Veraart J, Van Audekerke J, Couto J, Giugliano M, Wuyts K, Dedeurwaerdere S, Sijbers J, Mackie C, Ver Donck L, Steckler T, Van der Linden A. Subchronic memantine induced concurrent functional disconnectivity and altered ultra-structural tissue integrity in the rodent brain: revealed by multimodal MRI. Psychopharmacology (Berl) 2013; 227:479-91. [PMID: 23354531 DOI: 10.1007/s00213-013-2966-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 01/03/2013] [Indexed: 10/27/2022]
Abstract
BACKGROUND An effective NMDA antagonist imaging model may find key utility in advancing schizophrenia drug discovery research. We investigated effects of subchronic treatment with the NMDA antagonist memantine by using behavioural observation and multimodal MRI. METHODS Pharmacological MRI (phMRI) was used to map the neuroanatomical binding sites of memantine after acute and subchronic treatment. Resting state fMRI (rs-fMRI) and diffusion MRI were used to study the changes in functional connectivity (FC) and ultra-structural tissue integrity before and after subchronic memantine treatment. Further corroborating behavioural evidences were documented. RESULTS Dose-dependent phMRI activation was observed in the prelimbic cortex following acute doses of memantine. Subchronic treatment revealed significant effects in the hippocampus, cingulate, prelimbic and retrosplenial cortices. Decreases in FC amongst the hippocampal and frontal cortical structures (prelimbic, cingulate) were apparent through rs-fMRI investigation, indicating a loss of connectivity. Diffusion kurtosis MRI showed decreases in fractional anisotropy and mean diffusivity changes, suggesting ultra-structural changes in the hippocampus and cingulate cortex. Limited behavioural assessment suggested that memantine induced behavioural effects comparable to other NMDA antagonists as measured by locomotor hyperactivity and that the effects could be reversed by antipsychotic drugs. CONCLUSION Our findings substantiate the hypothesis that repeated NMDA receptor blockade with nonspecific, noncompetitive NMDA antagonists may lead to functional and ultra-structural alterations, particularly in the hippocampus and cingulate cortex. These changes may underlie the behavioural effects. Furthermore, the present findings underscore the utility and the translational potential of multimodal MR imaging and acute/subchronic memantine model in the search for novel disease-modifying treatments for schizophrenia.
Collapse
Affiliation(s)
- S Sekar
- Bio-Imaging Lab, Department of Biomedical Sciences, University of Antwerp, Campus Drie Eiken, D.UC.109, Universiteitsplein 1, 2610, Wilrijk, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Hikichi H, Kaku A, Karasawa JI, Chaki S. Stimulation of metabotropic glutamate (mGlu) 2 receptor and blockade of mGlu1 receptor improve social memory impairment elicited by MK-801 in rats. J Pharmacol Sci 2013; 122:10-6. [PMID: 23603933 DOI: 10.1254/jphs.13036fp] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Glutamatergic dysfunction has been implicated in psychiatric disorders such as schizophrenia. Both the stimulation of the metabotropic glutamate (mGlu) 2/3 receptor and the blockade of the mGlu1 receptor have been shown to be effective in a number of animal models of schizophrenia. However, the efficacy for social cognition, which is poorly managed by current medication, has not been fully addressed. The present study evaluated the effects of an mGlu2/3-receptor agonist and an mGlu1-receptor antagonist on social memory impairment in rats. Pretreatment with an mGlu2/3-receptor agonist, (-)-2-oxa-4-aminobicyclo[3.1.0]hexane-4,6-dicarboxylate (LY379268), or an mGlu1-receptor antagonist, (3,4-dihydro-2H-pyrano[2,3-b]quinolin-7-yl)-(cis-4-methoxycyclohexyl)-methanone (JNJ16259685), improved social memory impairment induced by 5R,10S-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine hydrogen maleate (MK-801) without affecting the social interactions. In addition, the intraperitoneal administration of an mGlu2-receptor potentiator, 3'-[[(2-cyclopentyl-2,3-dihydro-6,7-dimethyl-1-oxo-1H-inden-5-yl)oxy]methyl]-[1,1'-biphenyl]-4-carboxylic acid (BINA), also improved the MK-801-induced impairment of social memory, which was blocked by pretreatment with an mGlu2/3-receptor antagonist, (2S)-2-amino-2-[(1S,2S)-2-carboxycycloprop-1-yl]-3-(xanth-9-yl) propanoic acid (LY341495). These findings indicate that both the stimulation of the mGlu2 receptor and the inhibition of an mGlu1 receptor improve social memory impairment elicited by MK-801, and both manipulations could be effective approaches for the treatment of certain cognitive dysfunctions observed in schizophrenic patients.
Collapse
Affiliation(s)
- Hirohiko Hikichi
- Discovery Pharmacology I, Molecular Function and Pharmacology Laboratories, Taisho Pharmaceutical Co., Ltd., Japan
| | | | | | | |
Collapse
|
38
|
Agnoli L, Mainolfi P, Invernizzi RW, Carli M. Dopamine D1-like and D2-like receptors in the dorsal striatum control different aspects of attentional performance in the five-choice serial reaction time task under a condition of increased activity of corticostriatal inputs. Neuropsychopharmacology 2013; 38:701-14. [PMID: 23232445 PMCID: PMC3671986 DOI: 10.1038/npp.2012.236] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
We investigated the interaction between the corticostriatal glutamatergic afferents and dopamine D1-like and D2-like receptors in the dorsomedial striatum (dm-STR) in attention and executive response control in the five-choice serial reaction time (5-CSRT) task. The competitive NMDA receptor antagonist 3-(R)-2-carboxypiperazin-4-propyl-1-phosphonic acid (CPP) injected in the mPFC impaired accuracy and increased premature and perseverative responding, raising GLU, DA, and GABA release in the dm-STR. The D1-like antagonist SCH23390 injected in the dm-STR reversed the CPP-induced accuracy deficit but did not affect the increase in perseverative responding. In contrast, the D2-like antagonist haloperidol injected in the dm-STR reduced the CPP-induced increase in perseverative responding but not the accuracy deficit. The different roles of dorsal striatal D1-like and D2-like receptor were further supported by the finding that activation of D1-like receptor in the dm-STR by SKF38393 impaired accuracy but not perseverative responding while the D2-like agonist quinpirole injected in the dm-STR increased perseverative responding but did not affect accuracy. These findings suggest that integration of cortical information by D1-like receptors in the dm-STR is a key mechanism of the input selection process of attention while the integration of corticostriatal signals by D2-like receptors preserves the ability to switch from one act/response to the next in a complex motor sequence, thus providing for behavioral flexibility.
Collapse
Affiliation(s)
- Laura Agnoli
- Department of Neuroscience, Laboratory of Neurochemistry and Behavior, Istituto di Ricerche Farmacologiche ‘Mario Negri', via G. La Masa 19, Milano, Italy
| | - Pierangela Mainolfi
- Department of Neuroscience, Laboratory of Neurochemistry and Behavior, Istituto di Ricerche Farmacologiche ‘Mario Negri', via G. La Masa 19, Milano, Italy
| | - Roberto W Invernizzi
- Department of Neuroscience, Laboratory of Neurochemistry and Behavior, Istituto di Ricerche Farmacologiche ‘Mario Negri', via G. La Masa 19, Milano, Italy
| | - Mirjana Carli
- Department of Neuroscience, Laboratory of Neurochemistry and Behavior, Istituto di Ricerche Farmacologiche ‘Mario Negri', via G. La Masa 19, Milano, Italy,Department of Neuroscience, Laboratory of Neurochemistry and Behavior, Istituto di Ricerche Farmacologiche ‘Mario Negri', via G. La Masa 19, Milano 20156, Italy. Tel: +39 0239014466, Fax: +39 023546277, E-mail:
| |
Collapse
|
39
|
Becerra L, Upadhyay J, Chang PC, Bishop J, Anderson J, Baumgartner R, Schwarz AJ, Coimbra A, Wallin D, Nutile L, George E, Maier G, Sunkaraneni S, Iyengar S, Evelhoch JL, Bleakman D, Hargreaves R, Borsook D. Parallel buprenorphine phMRI responses in conscious rodents and healthy human subjects. J Pharmacol Exp Ther 2013; 345:41-51. [PMID: 23370795 DOI: 10.1124/jpet.112.201145] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Pharmacological magnetic resonance imaging (phMRI) is one method by which a drug's pharmacodynamic effects in the brain can be assessed. Although phMRI has been frequently used in preclinical and clinical settings, the extent to which a phMRI signature for a compound translates between rodents and humans has not been systematically examined. In the current investigation, we aimed to build on recent clinical work in which the functional response to 0.1 and 0.2 mg/70 kg i.v. buprenorphine (partial µ-opioid receptor agonist) was measured in healthy humans. Here, we measured the phMRI response to 0.04 and 0.1 mg/kg i.v. buprenorphine in conscious, naive rats to establish the parallelism of the phMRI signature of buprenorphine across species. PhMRI of 0.04 and 0.1 mg/kg i.v. buprenorphine yielded dose-dependent activation in a brain network composed of the somatosensory cortex, cingulate, insula, striatum, thalamus, periaqueductal gray, and cerebellum. Similar dose-dependent phMRI activation was observed in the human phMRI studies. These observations indicate an overall preservation of pharmacodynamic responses to buprenorphine between conscious, naive rodents and healthy human subjects, particularly in brain regions implicated in pain and analgesia. This investigation further demonstrates the usefulness of phMRI as a translational tool in neuroscience research that can provide mechanistic insight and guide dose selection in drug development.
Collapse
Affiliation(s)
- Lino Becerra
- Imaging Consortium for Drug Development, P.A.I.N. Group, Harvard Medical School, Children’s Hospital of Boston, Waltham, Massachusetts 02453, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Doyle OM, De Simoni S, Schwarz AJ, Brittain C, O'Daly OG, Williams SCR, Mehta MA. Quantifying the attenuation of the ketamine pharmacological magnetic resonance imaging response in humans: a validation using antipsychotic and glutamatergic agents. J Pharmacol Exp Ther 2013; 345:151-60. [PMID: 23370794 DOI: 10.1124/jpet.112.201665] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Ketamine acts as an N-methyl-D-aspartate receptor antagonist and evokes psychotomimetic symptoms resembling schizophrenia in healthy humans. Imaging markers of acute ketamine challenge have the potential to provide a powerful assay of novel therapies for psychiatric illness, although to date this assay has not been fully validated in humans. Pharmacological magnetic resonance imaging (phMRI) was conducted in a randomized, placebo-controlled crossover design in healthy volunteers. The study comprised a control and three ketamine infusion sessions, two of which included pretreatment with lamotrigine or risperidone, compounds hypothesized to reduce ketamine-induced glutamate release. The modulation of the ketamine phMRI response was investigated using univariate analysis of prespecified regions and a novel application of multivariate analysis across the whole-brain response. Lamotrigine and risperidone resulted in widespread attenuation of the ketamine-induced increases in signal, including the frontal and thalamic regions. A contrasting effect across both pretreatments was observed only in the subgenual prefrontal cortex, in which ketamine produced a reduction in signal. Multivariate techniques proved successful in both classifying ketamine from placebo (100%) and identifying the probability of scans belonging to the ketamine class (ketamine pretreated with placebo: 0.89). Following pretreatment, these predictive probabilities were reduced to 0.58 and 0.49 for lamotrigine and risperidone, respectively. We have provided clear demonstration of a ketamine phMRI response and its attenuation with both lamotrigine and risperidone. The analytical methodology used could be readily applied to investigate the mechanistic action of novel compounds relevant for psychiatric disorders such as schizophrenia and depression.
Collapse
Affiliation(s)
- O M Doyle
- King’s College London, Department of Neuroimaging, Institute of Psychiatry, London, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
41
|
Gozzi A, Agosta F, Massi M, Ciccocioppo R, Bifone A. Reduced limbic metabolism and fronto-cortical volume in rats vulnerable to alcohol addiction. Neuroimage 2012; 69:112-9. [PMID: 23261637 DOI: 10.1016/j.neuroimage.2012.12.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 12/07/2012] [Accepted: 12/10/2012] [Indexed: 11/25/2022] Open
Abstract
Alcohol abuse is associated with long-term reductions in fronto-cortical volume and limbic metabolism. However, an unanswered question in alcohol research is whether these alterations are the sole consequence of chronic alcohol use, or contain heritable contributions reflecting biological propensity toward ethanol addiction. Animal models of genetic predisposition to alcohol dependence can be used to investigate the role of inborn brain abnormalities in the aetiology of alcoholism. Here we used magnetic resonance imaging (MRI) in the Marchigian-Sardinian (msP) alcohol-preferring rats to assess the presence of inherited structural or functional brain alterations. Alcohol-naïve msP (N=22) and control rats (N=26) were subjected to basal cerebral blood volume (bCBV) mapping followed by voxel-based morphometry (VBM) of grey matter and tract-based spatial statistics mapping of white matter fractional anisotropy. msP rats exhibited significantly reduced bCBV, an established marker of resting brain function, in focal cortico-limbic and thalamic areas, together with reduced grey matter volume in the thalamus, ventral tegmental area, insular and cingulate cortex. No statistically significant differences in fractional anisotropy were observed between groups. These findings highlight the presence of inborn grey matter and metabolic abnormalities in alcohol-naïve msP rats, the localization and sign of which are remarkably similar to those mapped in abstinent alcoholics and subjects at high risk for alcohol dependence. Collectively, these results point for a significant role of heritable neurofunctional brain alterations in biological propensity toward ethanol addiction, and support the translational use of advanced imaging methods to describe the circuital determinants of vulnerability to drug addiction.
Collapse
Affiliation(s)
- Alessandro Gozzi
- Istituto Italiano di Tecnologia, Center for Nanotechnology Innovation, Pisa, & Center for Neuroscience and Cognitive Systems at UniTn, Rovereto, Italy.
| | | | | | | | | |
Collapse
|
42
|
Test-retest reliability of the BOLD pharmacological MRI response to ketamine in healthy volunteers. Neuroimage 2012; 64:75-90. [PMID: 23009959 DOI: 10.1016/j.neuroimage.2012.09.037] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 08/07/2012] [Accepted: 09/14/2012] [Indexed: 01/11/2023] Open
Abstract
The pharmacological MRI (phMRI) technique is being increasingly used in both pre-clinical and clinical models to investigate pharmacological effects on task-free brain function. Ketamine, an N-methyl-d-aspartate receptor (NMDAR) antagonist, induces a strong phMRI response and represents a promising pharmacological model to investigate the role of glutamatergic abnormalities in psychiatric symptomatology. The aim of this study was to assess whether the brain response to ketamine is reliable in order to validate ketamine phMRI as a mechanistic marker of glutamatergic dysfunction and to determine its utility in repeated measures designs to detect the modulatory effect of other drugs. Thus we assessed the test-retest reliability of the brain response to ketamine in healthy volunteers and identified an optimal modelling approach with reliability as our selection criterion. PhMRI data were collected from 10 healthy male participants, at rest, on two separate occasions. Subanaesthetic doses of I.V. ketamine infusion (target plasma levels 50 ng/mL and 75 ng/mL) were administered in both sessions. Test-retest reliability of the ketamine phMRI response was assessed voxel-wise and on pre-defined ROIs for a range of temporal design matrices including different combinations of nuisance regressors designed to model shape variance, linear drift and head motion. Effect sizes are also reported. All models showed a significant and widespread response to low-dose ketamine in predicted cerebral networks and as expected, increasing the number of model parameters improved model fit. Reliability of the predefined ROIs differed between the different models assessed. Using reliability as the selection criterion, a model capturing subject motion and linear drift performed the best across two sessions. The anatomical distribution of effects for all models was consistent with results of previous imaging studies in humans with BOLD signal increases in regions including midline cingulate and supracingulate cortex, thalamus, insula, anterior temporal lobe and ventrolateral prefrontal structures, and BOLD signal decreases in the subgenual cingulate cortex. This study represents the first investigation of the test-retest reliability of the BOLD phMRI response to acute ketamine challenge. All models tested were effective at describing the ketamine response although the design matrix associated with the highest reliability may represent a robust and well-characterised ketamine phMRI assay more suitable for repeated-measures designs. This ketamine assay is applicable as a model of neurotransmitter dysfunction suitable as a pharmacodynamic imaging tool to test and validate modulatory interventions, as a model of NMDA hypofunction in psychiatric disorders, and may be adapted to understand potential antidepressant and analgesic effects of NMDAR antagonists.
Collapse
|
43
|
Bifone A, Gozzi A. Neuromapping techniques in drug discovery: pharmacological MRI for the assessment of novel antipsychotics. Expert Opin Drug Discov 2012; 7:1071-82. [PMID: 22971143 DOI: 10.1517/17460441.2012.724057] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Treatment of psychiatric and neurological diseases represents a substantial unmet medical need, but the development of novel, effective and safe drugs is proving difficult. While substantial improvement over existing pharmacological agents is expected from new molecular targets emerging in the genomic era, the validation and exploitation of novel mechanisms of action is a lengthy and costly process. The use of neuroimaging techniques, and more specifically of functional and pharmacological magnetic resonance imaging (MRI), has been advocated as a powerful approach to this problem, providing translational biomarkers for the objective assessment of drug activity on brain function, and possibly surrogate markers of clinical response. AREAS COVERED The authors review the recent application of functional and pharmacological MRI (phMRI) in the study of novel treatments of psychosis based on glutamatergic mechanisms. Furthermore, they review contribution of functional imaging in the target validation and early assessment of drugs exploiting glutamatergic mechanisms as an example of potentially impactful exploitation of neuroimaging methods in drug discovery. EXPERT OPINION While functional neuroimaging methods may provide useful markers of drug activity and response to treatment, their translational potential, that is, their use to bridge animal and human investigations is seldom exploited. The application of phMRI in the study of novel antipsychotics based on glutamatergic mechanisms represents an example of functional neuroimaging as a powerful means to link preclinical and clinical research, thus providing a paradigm that may help expedite progression into the clinical phase of novel mechanisms for the treatment of psychiatric and neurological diseases.
Collapse
Affiliation(s)
- Angelo Bifone
- Istituto Italiano di Tecnologia, Center for Nanotechnology Innovation @NEST, Pisa, Italy.
| | | |
Collapse
|
44
|
Wellman PJ, Clifford PS, Rodriguez JA, Hughes S, Di Francesco C, Melotto S, Tessari M, Corsi M, Bifone A, Gozzi A. Brain reinforcement system function is ghrelin dependent: studies in the rat using pharmacological fMRI and intracranial self-stimulation. Addict Biol 2012; 17:908-19. [PMID: 22017465 DOI: 10.1111/j.1369-1600.2011.00392.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Ghrelin (GHR) is an orexigenic gut peptide that interacts with brain ghrelin receptors (GHR-Rs) to promote food intake. Recent research suggests that GHR acts as a modulator of motivated behavior, suggesting a direct influence of GHR on brain reinforcement circuits. In the present studies, we investigated the role of GHR and GHR-Rs in brain reinforcement function. Pharmacological magnetic resonance imaging was used to spatially resolve the functional activation produced by systemic administration of an orexigenic GHR dose. The imaging data revealed a focal activation of a network of subcortical structures that comprise brain reinforcement circuits-ventral tegmental area, lateral hypothalamus and nucleus accumbens. We next analyzed whether brain reinforcement circuits require functional GHR-Rs. To this purpose, wild-type (WT) or mutant rats sustaining N-ethyl-N-nitrosourea-induced knockout of GHR-Rs (GHR-R null rats) were implanted with stimulating electrodes aimed at the lateral hypothalamus, shaped to respond for intracranial self-stimulation (ICSS) and then tested using a rate-frequency procedure to examine ICSS response patterns. WT rats were readily shaped using stimulation intensities of 75 µA, whereas GHR-R null rats required 300 µA for ICSS shaping. No differences in rate-frequency curves were noted for WT rats at 75 µA and GHR-R null rats at 300 µA. When current intensity was lowered to 100 µA, GHR-R null rats did not respond for ICSS. Taken collectively, these data suggest that systemic GHR can activate mesolimbic dopaminergic areas, and highlight a facilitative role of GHR-Rs on the activity of brain reinforcement systems.
Collapse
Affiliation(s)
- Paul J Wellman
- Behavioral Neuroscience Program, Department of Psychology, Texas A&M University, College Station, TX 77843-4235, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Klomp A, Tremoleda JL, Schrantee A, Gsell W, Reneman L. The use of pharmacological-challenge fMRI in pre-clinical research: application to the 5-HT system. J Vis Exp 2012:3956. [PMID: 22565099 PMCID: PMC3466645 DOI: 10.3791/3956] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Pharmacological MRI (phMRI) is a new and promising method to study the effects of substances on brain function that can ultimately be used to unravel underlying neurobiological mechanisms behind drug action and neurotransmitter-related disorders, such as depression and ADHD. Like most of the imaging methods (PET, SPECT, CT) it represents a progress in the investigation of brain disorders and the related function of neurotransmitter pathways in a non-invasive way with respect of the overall neuronal connectivity. Moreover it also provides the ideal tool for translation to clinical investigations. MRI, while still behind in molecular imaging strategies compared to PET and SPECT, has the great advantage to have a high spatial resolution and no need for the injection of a contrast-agent or radio-labeled molecules, thereby avoiding the repetitive exposure to ionizing radiations. Functional MRI (fMRI) is extensively used in research and clinical setting, where it is generally combined with a psycho-motor task. phMRI is an adaptation of fMRI enabling the investigation of a specific neurotransmitter system, such as serotonin (5-HT), under physiological or pathological conditions following activation via administration of a specific challenging drug. The aim of the method described here is to assess brain 5-HT function in free-breathing animals. By challenging the 5-HT system while simultaneously acquiring functional MR images over time, the response of the brain to this challenge can be visualized. Several studies in animals have already demonstrated that drug-induced increases in extracellular levels of e.g. 5-HT (releasing agents, selective re-uptake blockers, etc) evoke region-specific changes in blood oxygenation level dependent (BOLD) MRI signals (signal due to a change of the oxygenated/deoxygenated hemoglobin levels occurring during brain activation through an increase of the blood supply to supply the oxygen and glucose to the demanding neurons) providing an index of neurotransmitter function. It has also been shown that these effects can be reversed by treatments that decrease 5-HT availability16,13,18,7. In adult rats, BOLD signal changes following acute SSRI administration have been described in several 5-HT related brain regions, i.e. cortical areas, hippocampus, hypothalamus and thalamus9,16,15. Stimulation of the 5-HT system and its response to this challenge can be thus used as a measure of its function in both animals and humans2,11.
Collapse
Affiliation(s)
- Anne Klomp
- Department of Radiology, Brain Imaging Center, Academic Medical Center Amsterdam
| | | | | | | | | |
Collapse
|
46
|
Fitzgerald PJ. The NMDA receptor may participate in widespread suppression of circuit level neural activity, in addition to a similarly prominent role in circuit level activation. Behav Brain Res 2012; 230:291-8. [DOI: 10.1016/j.bbr.2012.01.057] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 01/23/2012] [Accepted: 01/31/2012] [Indexed: 12/23/2022]
|
47
|
Pharmacologic magnetic resonance imaging (phMRI): imaging drug action in the brain. Neuroimage 2012; 62:1072-85. [PMID: 22495143 DOI: 10.1016/j.neuroimage.2012.03.075] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 03/23/2012] [Indexed: 02/01/2023] Open
Abstract
The technique of functional magnetic resonance (fMRI), using various cognitive, motor and sensory stimuli has led to a revolution in the ability to map brain function. Drugs can also be used as stimuli to elicit an hemodynamic change. Stimulation with a pharmaceutical has a number of very different consequences compared to user controllable stimuli, most importantly in the time course of stimulus and response that is not, in general, controllable by the experimenter. Therefore, this type of experiment has been termed pharmacologic MRI (phMRI). The use of a drug stimulus leads to a number of interesting possibilities compared to conventional fMRI. Using receptor specific ligands one can characterize brain circuitry specific to neurotransmitter systems. The possibility exists to measure parameters reflecting neurotransmitter release and binding associated with the pharmacokinetics and/or the pharmacodynamics of drugs. There is also the ability to measure up- and down-regulation of receptors in specific disease states. phMRI can be characterized as a molecular imaging technique using the natural hemodynamic transduction related to neuro-receptor stimulus. This provides a coupling mechanism with very high sensitivity that can rival positron emission tomography (PET) in some circumstances. The large numbers of molecules available, that do not require a radio-label, means that phMRI becomes a very useful tool for performing drug discovery. Data and arguments will be presented to show that phMRI can provide information on neuro-receptor signaling and function that complements the static picture generated by PET studies of receptor numbers and occupancies.
Collapse
|
48
|
Fell MJ, McKinzie DL, Monn JA, Svensson KA. Group II metabotropic glutamate receptor agonists and positive allosteric modulators as novel treatments for schizophrenia. Neuropharmacology 2012; 62:1473-83. [DOI: 10.1016/j.neuropharm.2011.06.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2011] [Revised: 06/07/2011] [Accepted: 06/08/2011] [Indexed: 10/18/2022]
|
49
|
Modulation of fronto-cortical activity by modafinil: a functional imaging and fos study in the rat. Neuropsychopharmacology 2012; 37:822-37. [PMID: 22048464 PMCID: PMC3260987 DOI: 10.1038/npp.2011.260] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Modafinil (MOD) is a wake-promoting drug with pro-cognitive properties. Despite its increasing use, the neuronal substrates of MOD action remain elusive. In particular, animal studies have highlighted a putative role of diencephalic areas as primary neuronal substrate of MOD action, with inconsistent evidence of recruitment of fronto-cortical areas despite the established pro-cognitive effects of the drug. Moreover, most animal studies have employed doses of MOD of limited clinical relevance. We used pharmacological magnetic resonance imaging (phMRI) in the anesthetized rat to map the circuitry activated by a MOD dose producing clinically relevant plasma exposure, as here ascertained by pharmacokinetic measurements. We observed prominent and sustained activation of the prefrontal and cingulate cortex, together with weaker but significant activation of the somatosensory cortex, medial thalamic domains, hippocampus, ventral striatum and dorsal raphe. Correlation analysis of phMRI data highlighted enhanced connectivity within a neural network including dopamine projections from the ventral tegmental area to the nucleus accumbens. The pro-arousing effect of MOD was assessed using electroencephalographic recording under anesthetic conditions comparable to those used for phMRI, together with the corresponding Fos immunoreactivity distribution. MOD produced electroencephalogram desynchronization, resulting in reduced delta and increased theta frequency bands, and a pattern of Fos induction largely consistent with the phMRI study. Altogether, these findings show that clinically relevant MOD doses can robustly activate fronto-cortical areas involved in higher cognitive functions and a network of pro-arousing areas, which provide a plausible substrate for the wake-promoting and pro-cognitive effects of the drug.
Collapse
|
50
|
A robust experimental protocol for pharmacological fMRI in rats and mice. J Neurosci Methods 2012; 204:9-18. [DOI: 10.1016/j.jneumeth.2011.10.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 10/20/2011] [Accepted: 10/22/2011] [Indexed: 01/03/2023]
|