1
|
Wang Z, Donahue EK, Guo Y, Renteln M, Petzinger GM, Jakowec MW, Holschneider DP. Exercise alters cortico-basal ganglia network metabolic connectivity: a mesoscopic level analysis informed by anatomic parcellation defined in the mouse brain connectome. Brain Struct Funct 2023; 228:1865-1884. [PMID: 37306809 PMCID: PMC10516800 DOI: 10.1007/s00429-023-02659-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 05/29/2023] [Indexed: 06/13/2023]
Abstract
The basal ganglia are important modulators of the cognitive and motor benefits of exercise. However, the neural networks underlying these benefits remain poorly understood. Our study systematically analyzed exercise-associated changes in metabolic connectivity in the cortico-basal ganglia-thalamic network during the performance of a new motor task, with regions-of-interest defined based on mesoscopic domains recently defined in the mouse brain structural connectome. Mice were trained on a motorized treadmill for six weeks or remained sedentary (control), thereafter undergoing [14C]-2-deoxyglucose metabolic brain mapping during wheel walking. Regional cerebral glucose uptake (rCGU) was analyzed in 3-dimensional brains reconstructed from autoradiographic brain sections using statistical parametric mapping. Metabolic connectivity was assessed by calculating inter-regional correlation of rCGU cross-sectionally across subjects within a group. Compared to controls, exercised animals showed broad decreases in rCGU in motor areas, but increases in limbic areas, as well as the visual and association cortices. In addition, exercised animals showed (i) increased positive metabolic connectivity within and between the motor cortex and caudoputamen (CP), (ii) newly emerged negative connectivity of the substantia nigra pars reticulata with the globus pallidus externus, and CP, and (iii) reduced connectivity of the prefrontal cortex (PFC). Increased metabolic connectivity in the motor circuit in the absence of increases in rCGU strongly suggests greater network efficiency, which is also supported by the reduced involvement of PFC-mediated cognitive control during the performance of a new motor task. Our study delineates exercise-associated changes in functional circuitry at the subregional level and provides a framework for understanding the effects of exercise on functions of the cortico-basal ganglia-thalamic network.
Collapse
Affiliation(s)
- Zhuo Wang
- Department of Psychiatry and Behavioral Sciences, University of Southern California, 1975 Zonal Avenue, KAM 400, MC9037, Los Angeles, California 90089-9037 USA
| | - Erin K. Donahue
- Graduate Program in Neurosciences, University of Southern California, Los Angeles, California USA
| | - Yumei Guo
- Department of Psychiatry and Behavioral Sciences, University of Southern California, 1975 Zonal Avenue, KAM 400, MC9037, Los Angeles, California 90089-9037 USA
| | - Michael Renteln
- Present Address: Department of Neurology, University of Southern California, Los Angeles, California USA
| | - Giselle M. Petzinger
- Graduate Program in Neurosciences, University of Southern California, Los Angeles, California USA
- Present Address: Department of Neurology, University of Southern California, Los Angeles, California USA
| | - Michael W. Jakowec
- Graduate Program in Neurosciences, University of Southern California, Los Angeles, California USA
- Present Address: Department of Neurology, University of Southern California, Los Angeles, California USA
| | - Daniel P. Holschneider
- Department of Psychiatry and Behavioral Sciences, University of Southern California, 1975 Zonal Avenue, KAM 400, MC9037, Los Angeles, California 90089-9037 USA
- Graduate Program in Neurosciences, University of Southern California, Los Angeles, California USA
- Present Address: Department of Neurology, University of Southern California, Los Angeles, California USA
- Department of Biomedical Engineering, University of Southern California, Los Angeles, California USA
| |
Collapse
|
2
|
Obrecht M, Zurbruegg S, Accart N, Lambert C, Doelemeyer A, Ledermann B, Beckmann N. Magnetic resonance imaging and ultrasound elastography in the context of preclinical pharmacological research: significance for the 3R principles. Front Pharmacol 2023; 14:1177421. [PMID: 37448960 PMCID: PMC10337591 DOI: 10.3389/fphar.2023.1177421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/16/2023] [Indexed: 07/18/2023] Open
Abstract
The 3Rs principles-reduction, refinement, replacement-are at the core of preclinical research within drug discovery, which still relies to a great extent on the availability of models of disease in animals. Minimizing their distress, reducing their number as well as searching for means to replace them in experimental studies are constant objectives in this area. Due to its non-invasive character in vivo imaging supports these efforts by enabling repeated longitudinal assessments in each animal which serves as its own control, thereby enabling to reduce considerably the animal utilization in the experiments. The repetitive monitoring of pathology progression and the effects of therapy becomes feasible by assessment of quantitative biomarkers. Moreover, imaging has translational prospects by facilitating the comparison of studies performed in small rodents and humans. Also, learnings from the clinic may be potentially back-translated to preclinical settings and therefore contribute to refining animal investigations. By concentrating on activities around the application of magnetic resonance imaging (MRI) and ultrasound elastography to small rodent models of disease, we aim to illustrate how in vivo imaging contributes primarily to reduction and refinement in the context of pharmacological research.
Collapse
Affiliation(s)
- Michael Obrecht
- Diseases of Aging and Regenerative Medicines, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Stefan Zurbruegg
- Neurosciences Department, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Nathalie Accart
- Diseases of Aging and Regenerative Medicines, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Christian Lambert
- Diseases of Aging and Regenerative Medicines, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Arno Doelemeyer
- Diseases of Aging and Regenerative Medicines, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Birgit Ledermann
- 3Rs Leader, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Nicolau Beckmann
- Diseases of Aging and Regenerative Medicines, Novartis Institutes for BioMedical Research, Basel, Switzerland
| |
Collapse
|
3
|
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: 5.3] [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
|
4
|
Zink N, Mückschel M, Beste C. Resting-state EEG Dynamics Reveals Differences in Network Organization and its Fluctuation between Frequency Bands. Neuroscience 2020; 453:43-56. [PMID: 33276088 DOI: 10.1016/j.neuroscience.2020.11.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 11/21/2020] [Accepted: 11/23/2020] [Indexed: 12/24/2022]
Abstract
Functional connectivity in EEG resting-state is not stable but fluctuates considerably. The aim of this study was to investigate how efficient information flows through a network, i.e. how resting-state EEG networks are organized and whether this organization it also subject to fluctuations. Differences of the network organization (small-worldness), degree of clustered connectivity, and path length as an indicator of how information is integrated into the network across time was compared between theta, alpha and beta bands. We show robust differences in network organization (small-worldness) between frequency bands. Fluctuations in network organization were larger in the theta, compared to the alpha and beta frequency. Variation in network organization and not the frequency of fluctuations differs between frequency bands. Furthermore, the degree of clustered connectivity and its modulation across time is the same across frequency bands, but the path length revealed the same modulatory pattern as the small-world metric. It is therefore the interplay of local processing efficiency and global information processing efficiency in the brain that fluctuates in a frequency-specific way. Properties of how information can be integrated is subject to fluctuations in a frequency-specific way in the resting-state. The possible relevance of these resting-state EEG properties is discussed including its clinical relevance.
Collapse
Affiliation(s)
- Nicolas Zink
- Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, United States; Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU, Dresden, Germany.
| | - Moritz Mückschel
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU, Dresden, Germany
| | - Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU, Dresden, Germany
| |
Collapse
|
5
|
Scuppa G, Tambalo S, Pfarr S, Sommer WH, Bifone A. Aberrant insular cortex connectivity in abstinent alcohol-dependent rats is reversed by dopamine D3 receptor blockade. Addict Biol 2020; 25:e12744. [PMID: 30907042 PMCID: PMC7187338 DOI: 10.1111/adb.12744] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/12/2019] [Accepted: 02/12/2019] [Indexed: 12/23/2022]
Abstract
A few studies have reported aberrant functional connectivity in alcoholic patients, but the specific neural circuits involved remain unknown. Moreover, it is unclear whether these alterations can be reversed upon treatment. Here, we used functional MRI to study resting state connectivity in rats following chronic intermittent exposure to ethanol. Further, we evaluated the effects of SB-277011-a, a selective dopamine D3 receptor antagonist, known to decrease ethanol consumption. Alcohol-dependent and control rats (N = 13/14 per group), 3 weeks into abstinence, were administered SB-277011-a or vehicle before fMRI sessions. Resting state connectivity networks were extracted by independent component analysis. A dual-regression analysis was performed using independent component maps as spatial regressors, and the effects of alcohol history and treatment on connectivity were assessed. A history of alcohol dependence caused widespread reduction of the internal coherence of components. Weaker correlation was also found between the insula cortex (IC) and cingulate cortices, key constituents of the salience network. Similarly, reduced connectivity was observed between a component comprising the anterior insular cortex, together with the caudate putamen (CPu-AntIns), and the posterior part of the IC. On the other hand, postdependent rats showed strengthened connectivity between salience and reward networks. In particular, higher connectivity was observed between insula and nucleus accumbens, between the ventral tegmental area and the cingulate cortex and between the VTA and CPu-AntIns. Interestingly, aberrant connectivity in postdependent rats was partially restored by acute administration of SB-277011-a, which, conversely, had no significant effects in naïve rats.
Collapse
Affiliation(s)
- Giulia Scuppa
- Center for Neuroscience and Cognitive SystemsIstituto Italiano di TecnologiaRoveretoItaly
| | - Stefano Tambalo
- Center for Neuroscience and Cognitive SystemsIstituto Italiano di TecnologiaRoveretoItaly
| | - Simone Pfarr
- Institute of Psychopharmacology, Central Institute of Mental HealthUniversity of HeidelbergMannheimGermany
| | - Wolfgang H. Sommer
- Institute of Psychopharmacology, Central Institute of Mental HealthUniversity of HeidelbergMannheimGermany
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental HealthUniversity of HeidelbergMannheimGermany
| | - Angelo Bifone
- Center for Neuroscience and Cognitive SystemsIstituto Italiano di TecnologiaRoveretoItaly
- Department of Molecular Biotechnology and Health SciencesUniversity of TorinoTorinoItaly
| |
Collapse
|
6
|
Holschneider DP, Wang Z, Guo Y, Sanford MT, Yeh J, Mao JJ, Zhang R, Rodriguez LV. Exercise modulates neuronal activation in the micturition circuit of chronically stressed rats: A multidisciplinary approach to the study of urologic chronic pelvic pain syndrome (MAPP) research network study. Physiol Behav 2020; 215:112796. [PMID: 31884113 PMCID: PMC7269603 DOI: 10.1016/j.physbeh.2019.112796] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 12/26/2019] [Accepted: 12/26/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Rats exposed to water avoidance stress (WAS) show increased urinary frequency, increased somatosensory nociceptive reflex responses, as well as altered brain responses to bladder distension, analogous to similar observations made in patients with urologic chronic pelvic pain syndrome (UCPPS). Exercise has been proposed as a potential treatment option for patients with chronic urinary frequency and urgency. We examined the effects of exercise on urinary voiding parameters and functional brain activation during bladder distension in rats exposed to WAS. METHODS Adult, female Wistar Kyoto rats were exposed to 10 days of WAS and thereafter randomized to either voluntary exercise for 3 weeks or sedentary groups. Voiding parameters were assessed at baseline, post-WAS, and weekly for 3 weeks. Thereafter, cerebral blood flow (CBF) mapping was performed during isotonic bladder distension (20 cm H2O) after intravenous bolus injection of [14C]-iodoantipyrine. Regional CBF was quantified in autoradiographs of brain slices and analyzed in 3-D reconstructed brains by statistical parametric mapping. Functional connectivity was examined between regions of the micturition circuit through interregional correlation analysis. RESULTS WAS exposure in sedentary animals (WAS/no-EX) increased voiding frequency and decreased urinary volumes per void. Exercise exposure in WAS animals (WAS/EX) resulted in a progressive decline in voiding frequency back to the baseline, as well as increased urinary volumes per void. Within the micturition circuit, WAS/EX compared to WAS/no-EX demonstrated a significantly lower rCBF response to passive bladder distension in Barrington's nucleus that is part of the spinobulbospinal voiding reflex, as well as in the periaqueductal gray (PAG) which modulates this reflex. Greater rCBF was noted in WAS/EX animals broadly across corticolimbic structures, including the cingulate, medial prefrontal cortex (prelimbic, infralimbic areas), insula, amygdala, and hypothalamus, which provide a 'top-down' decision point where micturition could be inhibited or triggered. WAS/EX showed a significantly greater positive brain functional connectivities compared to WAS/no-EX animals within regions of the extended reflex loop (PAG, Barrington's nucleus, intermediodorsal thalamic nucleus, pons), as well as within regions of the corticolimbic decision-making loop of the micturition circuit, with a strikingly negative correlation between these pathways. Urinary frequency was positively correlated with rCBF in the pons, and negatively correlated with rCBF in the cingulate cortex. CONCLUSION Our results suggest that chronic voluntary exercise may decrease urinary frequency at two points of control in the micturition circuit. During the urine storage phase, it may diminish the influence of the reflex micturition circuit itself, and/or it may increase corticolimbic control of voiding. Exercise may be an effective adjunct therapeutic intervention for modifying the urinary symptoms in patients with UCPPS.
Collapse
Affiliation(s)
- Daniel P Holschneider
- Departments of Psychiatry and the Behavioral Sciences, Los Angeles, CA, United States.
| | - Zhuo Wang
- Departments of Psychiatry and the Behavioral Sciences, Los Angeles, CA, United States
| | - Yumei Guo
- Departments of Psychiatry and the Behavioral Sciences, Los Angeles, CA, United States
| | - Melissa T Sanford
- Urology at the University of Southern California, Los Angeles, CA, United States
| | - Jihchao Yeh
- Urology at the University of Southern California, Los Angeles, CA, United States
| | - Jackie J Mao
- Urology at the University of Southern California, Los Angeles, CA, United States
| | - Rong Zhang
- Urology at the University of Southern California, Los Angeles, CA, United States
| | - Larissa V Rodriguez
- Urology at the University of Southern California, Los Angeles, CA, United States.
| |
Collapse
|
7
|
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: 16.5] [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
|
8
|
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: 3.8] [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
|
9
|
Holschneider DP, Givrad TK, Yang J, Stewart SB, Francis SR, Wang Z, Maarek J. Cerebral perfusion mapping during retrieval of spatial memory in rats. Behav Brain Res 2019; 375:112116. [PMID: 31377254 DOI: 10.1016/j.bbr.2019.112116] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/26/2019] [Accepted: 07/26/2019] [Indexed: 12/24/2022]
Abstract
Studies of brain functional activation during spatial navigation using electrophysiology and immediate-early gene responses have typically targeted a limited number of brain regions. Our study provides the first whole brain analysis of cerebral activation during retrieval of spatial memory in the freely-moving rat. Rats (LEARNERS) were trained in the Barnes maze, an allocentric spatial navigation task, while CONTROLS received passive exposure. After 19 days, functional brain mapping was performed during recall by bolus intravenous injection of [14C]-iodoantipyrine using a novel subcutaneous minipump triggered by remote activation. Regional cerebral blood flow (rCBF)-related tissue radioactivity was analyzed by statistical parametric mapping from autoradiographic images of the three-dimensionally reconstructed brains. Functional connectivity was examined between regions of the spatial navigation circuit through interregional correlation analysis. Significant rCBF increases were noted in LEARNERS compared to CONTROLS broadly across the spatial navigation circuit, including the hippocampus (anterior dorsal CA1, posterior ventral CA1-3), subiculum, thalamus, striatum, medial septum, cerebral cortex, with decreases noted in the mammillary nucleus, amygdala and insula. LEARNERS showed a significantly greater positive correlation of rCBF of the ventral hippocampus with retrosplenial, lateral orbital, parietal and primary visual cortex, and a significantly more negative correlation with the mammillary nucleus, amygdala, posterior entorhinal cortex, and anterior thalamic nucleus. The complex sensory component of the spatial navigation task was underscored by broad activation across visual, somatosensory, olfactory, auditory and vestibular circuits which was enhanced in LEARNERS. Brain mapping facilitated by an implantable minipump represents a powerful tool for evaluation of mammalian behaviors dependent on locomotion.
Collapse
Affiliation(s)
- D P Holschneider
- Dept. of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, United States; Dept. of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, United States; Viterbi School of Engineering, Dept. of Biomedical Engineering, Los Angeles, CA, 90033, United States.
| | - T K Givrad
- Viterbi School of Engineering, Dept. of Biomedical Engineering, Los Angeles, CA, 90033, United States
| | - J Yang
- Dept. of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, United States
| | - S B Stewart
- Dept. of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, United States
| | - S R Francis
- Dept. of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, United States
| | - Z Wang
- Dept. of Psychiatry and the Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90089, United States
| | - Jmi Maarek
- Viterbi School of Engineering, Dept. of Biomedical Engineering, Los Angeles, CA, 90033, United States
| |
Collapse
|
10
|
Central Noradrenergic Agonists in the Treatment of Ischemic Stroke-an Overview. Transl Stroke Res 2019; 11:165-184. [PMID: 31327133 DOI: 10.1007/s12975-019-00718-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 02/03/2023]
Abstract
Ischemic stroke is the leading cause of morbidity and mortality with a significant health burden worldwide and few treatment options. Among the short- and long-term effects of ischemic stroke is the cardiovascular sympathetic autonomic dysfunction, presented in part as the by-product of the ischemic damage to the noradrenergic centers of the brain. Unlike high levels in the plasma, the brain may face suboptimal levels of norepinephrine (NE), with adverse effects on the clinical and functional outcomes of ischemic stroke. The intravenous administration of NE and other sympathomimetic agents, in an attempt to increase cerebral perfusion pressure, often aggravates the ischemia-induced rise in blood pressure (BP) with life-threatening consequences for stroke patients, the majority of whom present with hypertension at the time of admission. Unlike the systemic administration, the central administration of NE reduces BP while exerting anti-inflammatory and neuroprotective effects. These characteristics of centrally administered NE, combined with the short latency of response, make it an ideal candidate for use in the acute phase of stroke, followed by the use of centrally acting noradrenergic agonists, such as NE reuptake inhibitors and B2-adrenergic receptor agonists for stroke rehabilitation. In addition, a number of nonpharmacological strategies, such as transcutaneous vagus nerve stimulation (tVNS) and trigeminal nerve stimulation (TNS), have the potential to enhance the central noradrenergic functional activities and improve stroke clinical outcomes. Many factors could influence the efficacy of the noradrenergic treatment in stroke patients. These factors include the type of the noradrenergic agent; the dose, frequency, and duration of administration; the timing of administration in relation to the acute event; and the site and characteristics of the ischemic lesions. Having this knowledge, combined with the better understanding of the regulation of noradrenergic receptors in different parts of the brain, would pave the path for the successful use of the centrally acting noradrenergic agents in the management of ischemic stroke.
Collapse
|
11
|
Tollens F, Gass N, Becker R, Schwarz AJ, Risterucci C, Künnecke B, Lebhardt P, Reinwald J, Sack M, Weber-Fahr W, Meyer-Lindenberg A, Sartorius A. The affinity of antipsychotic drugs to dopamine and serotonin 5-HT 2 receptors determines their effects on prefrontal-striatal functional connectivity. Eur Neuropsychopharmacol 2018; 28:1035-1046. [PMID: 30006253 DOI: 10.1016/j.euroneuro.2018.05.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 03/07/2018] [Accepted: 05/17/2018] [Indexed: 12/31/2022]
Abstract
One of the major challenges of cross-species translation in psychiatry is the identification of quantifiable brain phenotypes linked to drug efficacy and/or side effects. A measure that has received increasing interest is the effect of antipsychotic drugs on resting-state functional connectivity (FC) in magnetic resonance imaging. However, quantitative comparisons of antipsychotic drug-induced alterations of FC patterns are missing. Consideration of receptor binding affinities provides a means for the effects of antipsychotic drugs on extended brain networks to be related directly to their molecular mechanism of action. Therefore, we examined the relationship between the affinities of three second-generation antipsychotics (amisulpride, risperidone and olanzapine) to dopamine and serotonin receptors and FC patterns related to the prefrontal cortex (PFC) and striatum in Sprague-Dawley rats. FC of the relevant regions was quantified by correlation coefficients and local network properties. Each drug group (32 animals per group) was subdivided into three dose groups and a vehicle control group. A linear relationship was discovered for the mid-dose of antipsychotic compounds, with stronger affinity to serotonin 5-HT2A, 5-HT2C and 5-HT1A receptors and decreased affinity to D3 receptors associated with increased prefrontal-striatal FC (p = 0.0004, r² = 0.46; p = 0.004, r² = 0.33; p = 0.002, r² = 0.37; p = 0.02, r² = 0.22, respectively). Interestingly, no correlation was observed for the low and high dose groups, and for D2 receptors. Our results indicate that drug-induced FC patterns may be linked to antipsychotic mechanism of action on the molecular level and suggest the technique's value for drug development, especially if our results are extended to a larger number of antipsychotics.
Collapse
Affiliation(s)
- F Tollens
- Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - N Gass
- Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - R Becker
- Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - A J Schwarz
- Eli Lilly and Company, Indianapolis, IN 46285, USA; Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN 47405, USA; Department of Radiological and Imaging Sciences, Indiana University School of Medicine, Indiana University - Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - C Risterucci
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - B Künnecke
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - P Lebhardt
- Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - J Reinwald
- Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany; Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - M Sack
- Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - W Weber-Fahr
- Department of Neuroimaging, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - A Meyer-Lindenberg
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - A Sartorius
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
12
|
Faraone SV. The pharmacology of amphetamine and methylphenidate: Relevance to the neurobiology of attention-deficit/hyperactivity disorder and other psychiatric comorbidities. Neurosci Biobehav Rev 2018; 87:255-270. [PMID: 29428394 DOI: 10.1016/j.neubiorev.2018.02.001] [Citation(s) in RCA: 311] [Impact Index Per Article: 51.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/25/2018] [Accepted: 02/05/2018] [Indexed: 12/20/2022]
Abstract
Psychostimulants, including amphetamines and methylphenidate, are first-line pharmacotherapies for individuals with attention-deficit/hyperactivity disorder (ADHD). This review aims to educate physicians regarding differences in pharmacology and mechanisms of action between amphetamine and methylphenidate, thus enhancing physician understanding of psychostimulants and their use in managing individuals with ADHD who may have comorbid psychiatric conditions. A systematic literature review of PubMed was conducted in April 2017, focusing on cellular- and brain system-level effects of amphetamine and methylphenidate. The primary pharmacologic effect of both amphetamine and methylphenidate is to increase central dopamine and norepinephrine activity, which impacts executive and attentional function. Amphetamine actions include dopamine and norepinephrine transporter inhibition, vesicular monoamine transporter 2 (VMAT-2) inhibition, and monoamine oxidase activity inhibition. Methylphenidate actions include dopamine and norepinephrine transporter inhibition, agonist activity at the serotonin type 1A receptor, and redistribution of the VMAT-2. There is also evidence for interactions with glutamate and opioid systems. Clinical implications of these actions in individuals with ADHD with comorbid depression, anxiety, substance use disorder, and sleep disturbances are discussed.
Collapse
Affiliation(s)
- Stephen V Faraone
- Departments of Psychiatry and of Neuroscience and Physiology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States; K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway.
| |
Collapse
|
13
|
Effects of dexamphetamine-induced dopamine release on resting-state network connectivity in recreational amphetamine users and healthy controls. Brain Imaging Behav 2017; 10:548-58. [PMID: 26149196 PMCID: PMC4908160 DOI: 10.1007/s11682-015-9419-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Dexamphetamine (dAMPH) is not only used for the treatment of attention deficit hyperactivity disorder (ADHD), but also as a recreational drug. Acutely, dAMPH induces release of predominantly dopamine (DA) in the striatum, and in the cortex both DA and noradrenaline. Recent animal studies have shown that chronic dAMPH administration can induce changes in the DA system following long-term exposure, as evidenced by reductions in DA transporters, D2/3 receptors and endogenous DA levels. However, only a limited number of studies have investigated the effects of dAMPH in the human brain. We used a combination of resting-state functional magnetic resonance imaging (rs-fMRI) and [(123)I]IBZM single-photon emission computed tomography (SPECT) (to assess baseline D2/3 receptor binding and DA release) in 15 recreational AMPH users and 20 matched healthy controls to investigate the short-, and long-term effects of AMPH before and after an acute intravenous challenge with dAMPH. We found that acute dAMPH administration reduced functional connectivity in the cortico-striatal-thalamic network. dAMPH-induced DA release, but not DA D2/3 receptor binding, was positively associated with connectivity changes in this network. In addition, acute dAMPH reduced connectivity in default mode networks and salience-executive-networks networks in both groups. In contrast to our hypothesis, no significant group differences were found in any of the rs-fMRI networks investigated, possibly due to lack of sensitivity or compensatory mechanisms. Our findings thus support the use of ICA-based resting-state functional connectivity as a tool to investigate acute, but not chronic, alterations induced by dAMPH on dopaminergic processing in the striatum.
Collapse
|
14
|
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: 1.0] [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
|
15
|
Khalili-Mahani N, Rombouts SARB, van Osch MJP, Duff EP, Carbonell F, Nickerson LD, Becerra L, Dahan A, Evans AC, Soucy JP, Wise R, Zijdenbos AP, van Gerven JM. Biomarkers, designs, and interpretations of resting-state fMRI in translational pharmacological research: A review of state-of-the-Art, challenges, and opportunities for studying brain chemistry. Hum Brain Mapp 2017; 38:2276-2325. [PMID: 28145075 DOI: 10.1002/hbm.23516] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 11/21/2016] [Accepted: 01/04/2017] [Indexed: 12/11/2022] Open
Abstract
A decade of research and development in resting-state functional MRI (RSfMRI) has opened new translational and clinical research frontiers. This review aims to bridge between technical and clinical researchers who seek reliable neuroimaging biomarkers for studying drug interactions with the brain. About 85 pharma-RSfMRI studies using BOLD signal (75% of all) or arterial spin labeling (ASL) were surveyed to investigate the acute effects of psychoactive drugs. Experimental designs and objectives include drug fingerprinting dose-response evaluation, biomarker validation and calibration, and translational studies. Common biomarkers in these studies include functional connectivity, graph metrics, cerebral blood flow and the amplitude and spectrum of BOLD fluctuations. Overall, RSfMRI-derived biomarkers seem to be sensitive to spatiotemporal dynamics of drug interactions with the brain. However, drugs cause both central and peripheral effects, thus exacerbate difficulties related to biological confounds, structured noise from motion and physiological confounds, as well as modeling and inference testing. Currently, these issues are not well explored, and heterogeneities in experimental design, data acquisition and preprocessing make comparative or meta-analysis of existing reports impossible. A unifying collaborative framework for data-sharing and data-mining is thus necessary for investigating the commonalities and differences in biomarker sensitivity and specificity, and establishing guidelines. Multimodal datasets including sham-placebo or active control sessions and repeated measurements of various psychometric, physiological, metabolic and neuroimaging phenotypes are essential for pharmacokinetic/pharmacodynamic modeling and interpretation of the findings. We provide a list of basic minimum and advanced options that can be considered in design and analyses of future pharma-RSfMRI studies. Hum Brain Mapp 38:2276-2325, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Najmeh Khalili-Mahani
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, McGill University, Montreal, Canada.,PERFORM Centre, Concordia University, Montreal, Canada
| | - Serge A R B Rombouts
- Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands.,Institute of Psychology and Leiden Institute for Brain and Cognition, Leiden University, Leiden, The Netherlands
| | | | - Eugene P Duff
- Institute of Psychology and Leiden Institute for Brain and Cognition, Leiden University, Leiden, The Netherlands.,Oxford Centre for Functional MRI of the Brain, Oxford University, Oxford, United Kingdom
| | | | - Lisa D Nickerson
- McLean Hospital, Belmont, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - Lino Becerra
- Center for Pain and the Brain, Harvard Medical School & Boston Children's Hospital, Boston, Massachusetts
| | - Albert Dahan
- Department of Anesthesiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Alan C Evans
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, McGill University, Montreal, Canada.,McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Jean-Paul Soucy
- PERFORM Centre, Concordia University, Montreal, Canada.,McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Richard Wise
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, United Kingdom
| | - Alex P Zijdenbos
- McGill Centre for Integrative Neuroscience, Montreal Neurological Institute, McGill University, Montreal, Canada.,Biospective Inc, Montreal, Quebec, Canada
| | - Joop M van Gerven
- Centre for Human Drug Research, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
16
|
Bär KJ, de la Cruz F, Schumann A, Koehler S, Sauer H, Critchley H, Wagner G. Functional connectivity and network analysis of midbrain and brainstem nuclei. Neuroimage 2016; 134:53-63. [PMID: 27046112 DOI: 10.1016/j.neuroimage.2016.03.071] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 03/22/2016] [Accepted: 03/26/2016] [Indexed: 12/23/2022] Open
|
17
|
Jonckers E, Shah D, Hamaide J, Verhoye M, Van der Linden A. The power of using functional fMRI on small rodents to study brain pharmacology and disease. Front Pharmacol 2015; 6:231. [PMID: 26539115 PMCID: PMC4612660 DOI: 10.3389/fphar.2015.00231] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/28/2015] [Indexed: 12/23/2022] Open
Abstract
Functional magnetic resonance imaging (fMRI) is an excellent tool to study the effect of pharmacological modulations on brain function in a non-invasive and longitudinal manner. We introduce several blood oxygenation level dependent (BOLD) fMRI techniques, including resting state (rsfMRI), stimulus-evoked (st-fMRI), and pharmacological MRI (phMRI). Respectively, these techniques permit the assessment of functional connectivity during rest as well as brain activation triggered by sensory stimulation and/or a pharmacological challenge. The first part of this review describes the physiological basis of BOLD fMRI and the hemodynamic response on which the MRI contrast is based. Specific emphasis goes to possible effects of anesthesia and the animal’s physiological conditions on neural activity and the hemodynamic response. The second part of this review describes applications of the aforementioned techniques in pharmacologically induced, as well as in traumatic and transgenic disease models and illustrates how multiple fMRI methods can be applied successfully to evaluate different aspects of a specific disorder. For example, fMRI techniques can be used to pinpoint the neural substrate of a disease beyond previously defined hypothesis-driven regions-of-interest. In addition, fMRI techniques allow one to dissect how specific modifications (e.g., treatment, lesion etc.) modulate the functioning of specific brain areas (st-fMRI, phMRI) and how functional connectivity (rsfMRI) between several brain regions is affected, both in acute and extended time frames. Furthermore, fMRI techniques can be used to assess/explore the efficacy of novel treatments in depth, both in fundamental research as well as in preclinical settings. In conclusion, by describing several exemplary studies, we aim to highlight the advantages of functional MRI in exploring the acute and long-term effects of pharmacological substances and/or pathology on brain functioning along with several methodological considerations.
Collapse
Affiliation(s)
- Elisabeth Jonckers
- Bio-Imaging Lab, Department of Biomedical Sciences, University of Antwerp Antwerp, Belgium
| | - Disha Shah
- Bio-Imaging Lab, Department of Biomedical Sciences, University of Antwerp Antwerp, Belgium
| | - Julie Hamaide
- Bio-Imaging Lab, Department of Biomedical Sciences, University of Antwerp Antwerp, Belgium
| | - Marleen Verhoye
- Bio-Imaging Lab, Department of Biomedical Sciences, University of Antwerp Antwerp, Belgium
| | - Annemie Van der Linden
- Bio-Imaging Lab, Department of Biomedical Sciences, University of Antwerp Antwerp, Belgium
| |
Collapse
|
18
|
Sárvári M, Deli L, Kocsis P, Márk L, Maász G, Hrabovszky E, Kalló I, Gajári D, Vastagh C, Sümegi B, Tihanyi K, Liposits Z. Estradiol and isotype-selective estrogen receptor agonists modulate the mesocortical dopaminergic system in gonadectomized female rats. Brain Res 2014; 1583:1-11. [DOI: 10.1016/j.brainres.2014.06.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 06/18/2014] [Accepted: 06/19/2014] [Indexed: 01/08/2023]
|
19
|
Smucny J, Wylie KP, Tregellas JR. Functional magnetic resonance imaging of intrinsic brain networks for translational drug discovery. Trends Pharmacol Sci 2014; 35:397-403. [PMID: 24906509 DOI: 10.1016/j.tips.2014.05.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 04/30/2014] [Accepted: 05/05/2014] [Indexed: 01/23/2023]
Abstract
Developing translational biomarkers is a priority for psychiatry research. Task-independent functional brain imaging is a relatively novel technique that allows examination of the brain's intrinsic networks, defined as functionally and (often) structurally connected populations of neurons whose properties reflect fundamental neurobiological organizational principles of the central nervous system. The ability to study the activity and organization of these networks has opened a promising new avenue for translational investigation, because they can be analogously examined across species and disease states. Interestingly, imaging studies have revealed shared spatial and functional characteristics of the intrinsic network architecture of the brain across species, including mice, rats, non-human primates, and humans. Using schizophrenia as an example, we show how intrinsic networks may show similar abnormalities in human diseases and animal models of these diseases, supporting their use as biomarkers in drug development.
Collapse
Affiliation(s)
- Jason Smucny
- Research Service, Denver VA Medical Center, Denver, CO, USA; Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Neuroscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Korey P Wylie
- Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jason R Tregellas
- Research Service, Denver VA Medical Center, Denver, CO, USA; Department of Psychiatry, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Neuroscience Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
20
|
Byun NE, Grannan M, Bubser M, Barry RL, Thompson A, Rosanelli J, Gowrishankar R, Kelm ND, Damon S, Bridges TM, Melancon BJ, Tarr JC, Brogan JT, Avison MJ, Deutch AY, Wess J, Wood MR, Lindsley CW, Gore JC, Conn PJ, Jones CK. Antipsychotic drug-like effects of the selective M4 muscarinic acetylcholine receptor positive allosteric modulator VU0152100. Neuropsychopharmacology 2014; 39:1578-93. [PMID: 24442096 PMCID: PMC4023154 DOI: 10.1038/npp.2014.2] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 12/06/2013] [Accepted: 12/12/2013] [Indexed: 12/18/2022]
Abstract
Accumulating evidence suggests that selective M4 muscarinic acetylcholine receptor (mAChR) activators may offer a novel strategy for the treatment of psychosis. However, previous efforts to develop selective M4 activators were unsuccessful because of the lack of M4 mAChR subtype specificity and off-target muscarinic adverse effects. We recently developed VU0152100, a highly selective M4 positive allosteric modulator (PAM) that exerts central effects after systemic administration. We now report that VU0152100 dose-dependently reverses amphetamine-induced hyperlocomotion in rats and wild-type mice, but not in M4 KO mice. VU0152100 also blocks amphetamine-induced disruption of the acquisition of contextual fear conditioning and prepulse inhibition of the acoustic startle reflex. These effects were observed at doses that do not produce catalepsy or peripheral adverse effects associated with non-selective mAChR agonists. To further understand the effects of selective potentiation of M4 on region-specific brain activation, VU0152100 alone and in combination with amphetamine were evaluated using pharmacologic magnetic resonance imaging (phMRI). Key neural substrates of M4-mediated modulation of the amphetamine response included the nucleus accumbens (NAS), caudate-putamen (CP), hippocampus, and medial thalamus. Functional connectivity analysis of phMRI data, specifically assessing correlations in activation between regions, revealed several brain networks involved in the M4 modulation of amphetamine-induced brain activation, including the NAS and retrosplenial cortex with motor cortex, hippocampus, and medial thalamus. Using in vivo microdialysis, we found that VU0152100 reversed amphetamine-induced increases in extracellular dopamine levels in NAS and CP. The present data are consistent with an antipsychotic drug-like profile of activity for VU0152100. Taken together, these data support the development of selective M4 PAMs as a new approach to the treatment of psychosis and cognitive impairments associated with psychiatric disorders such as schizophrenia.
Collapse
Affiliation(s)
- Nellie E Byun
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA,Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael Grannan
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael Bubser
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Robert L Barry
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Analisa Thompson
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John Rosanelli
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Raajaram Gowrishankar
- Neuroscience Graduate Program, Vanderbilt University, Nashville, TN, USA,Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA,Vanderbilt International Scholars Program, Vanderbilt University, Nashville, TN, USA
| | - Nathaniel D Kelm
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Stephen Damon
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Thomas M Bridges
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Chemistry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bruce J Melancon
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James C Tarr
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John T Brogan
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Chemistry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Malcolm J Avison
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA,Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ariel Y Deutch
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jürgen Wess
- Molecular Signaling Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael R Wood
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Chemistry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Craig W Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Chemistry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John C Gore
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - P Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Carrie K Jones
- Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA,Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical Center, 418B Preston Research Building, Nashville, TN 37232, USA, Tel: +1 615 343 4337, Fax: +1 615 343 3088, E-mail:
| |
Collapse
|
21
|
Lu H, Stein EA. Resting state functional connectivity: its physiological basis and application in neuropharmacology. Neuropharmacology 2013; 84:79-89. [PMID: 24012656 DOI: 10.1016/j.neuropharm.2013.08.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 07/14/2013] [Accepted: 08/21/2013] [Indexed: 11/29/2022]
Abstract
Brain structures do not work in isolation; they work in concert to produce sensory perception, motivation and behavior. Systems-level network activity can be investigated by resting state magnetic resonance imaging (rsMRI), an emerging neuroimaging technique that assesses the synchrony of the brain's ongoing spontaneous activity. Converging evidence reveals that rsMRI is able to consistently identify distinct spatiotemporal patterns of large-scale brain networks. Dysregulation within and between these networks has been implicated in a number of neurodegenerative and neuropsychiatric disorders, including Alzheimer's disease and drug addiction. Despite wide application of this approach in systems neuroscience, the physiological basis of these fluctuations remains incompletely understood. Here we review physiological studies in electrical, metabolic and hemodynamic fluctuations that are most pertinent to the rsMRI signal. We also review recent applications to neuropharmacology - specifically drug effects on resting state fluctuations. We speculate that the mechanisms governing spontaneous fluctuations in regional oxygenation availability likely give rise to the observed rsMRI signal. We conclude by identifying several open questions surrounding this technique. This article is part of the Special Issue Section entitled 'Neuroimaging in Neuropharmacology'.
Collapse
Affiliation(s)
- Hanbing Lu
- Neuroimaging Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, USA.
| | - Elliot A Stein
- Neuroimaging Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, USA
| |
Collapse
|
22
|
Matthews PM, Coatney R, Alsaid H, Jucker B, Ashworth S, Parker C, Changani K. Technologies: preclinical imaging for drug development. DRUG DISCOVERY TODAY. TECHNOLOGIES 2013; 10:e343-e350. [PMID: 24050130 DOI: 10.1016/j.ddtec.2012.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Preclinical imaging with magnetic resonance imaging (MRI), computerised tomography (CT), ultrasound (US), positron emission tomography (PET) or single-photon emission computed tomography (SPECT) enable non-invasive measures of tissue structure, function or metabolism in vivo. The technologies can add value to preclinical studies by enabling dynamic pharmacological observations on the same animal and because of possibilities for relatively direct clinical translation. Potential benefits from the application of preclinical imaging should be considered routinely in drug development.
Collapse
|
23
|
Jonckers E, Van der Linden A, Verhoye M. Functional magnetic resonance imaging in rodents: an unique tool to study in vivo pharmacologic neuromodulation. Curr Opin Pharmacol 2013; 13:813-20. [PMID: 23856429 DOI: 10.1016/j.coph.2013.06.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 06/25/2013] [Accepted: 06/25/2013] [Indexed: 10/26/2022]
Abstract
When new compounds targeting the brain are developed, it is important to assess both the acute and chronic effects on brain functioning. This can be done non-invasively using a technique called functional magnetic resonance imaging (fMRI). This review discusses the possibilities of both stimulation-based and resting state fMRI to study pharmacological modulations of the rodent brain. Moreover, attention is given to the use of anesthetics which could importantly influence the outcome of both techniques.
Collapse
|
24
|
Pharmacological imaging as a tool to visualise dopaminergic neurotoxicity. Neuropharmacology 2013; 84:159-69. [PMID: 23851258 DOI: 10.1016/j.neuropharm.2013.06.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 06/06/2013] [Accepted: 06/27/2013] [Indexed: 11/20/2022]
Abstract
Dopamine abnormalities underlie a wide variety of psychopathologies, including ADHD and schizophrenia. A new imaging technique, pharmacological magnetic resonance imaging (phMRI), is a promising non-invasive technique to visualize the dopaminergic system in the brain. In this review we explore the clinical potential of phMRI in detecting dopamine dysfunction or neurotoxicity, assess its strengths and weaknesses and identify directions for future research. Preclinically, phMRI is able to detect severe dopaminergic abnormalities quite similar to conventional techniques such as PET and SPECT. phMRI benefits from its high spatial resolution and the possibility to visualize both local and downstream effects of dopaminergic neurotransmission. In addition, it allows for repeated measurements and assessments in vulnerable populations. The major challenge is the complex interpretation of phMRI results. Future studies in patients with dopaminergic abnormalities need to confirm the currently reviewed preclinical findings to validate the technique in a clinical setting. Eventually, based on the current review we expect that phMRI can be of use in a clinical setting involving vulnerable populations (such as children and adolescents) for diagnosis and monitoring treatment efficacy. This article is part of the Special Issue Section entitled 'Neuroimaging in Neuropharmacology'.
Collapse
|
25
|
Razoux F, Baltes C, Mueggler T, Seuwen A, Russig H, Mansuy I, Rudin M. Functional MRI to assess alterations of functional networks in response to pharmacological or genetic manipulations of the serotonergic system in mice. Neuroimage 2013; 74:326-36. [DOI: 10.1016/j.neuroimage.2013.02.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 01/17/2013] [Accepted: 02/11/2013] [Indexed: 01/21/2023] Open
|
26
|
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: 1.5] [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
|
27
|
Heidbreder C. Rationale in support of the use of selective dopamine D₃ receptor antagonists for the pharmacotherapeutic management of substance use disorders. Naunyn Schmiedebergs Arch Pharmacol 2012; 386:167-76. [PMID: 23104235 DOI: 10.1007/s00210-012-0803-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 10/15/2012] [Indexed: 10/27/2022]
Abstract
Growing evidence indicates that dopamine (DA) D(3) receptors are involved in the control of drug-seeking behavior and may play an important role in the pathophysiology of substance use disorders. First, DA D(3) receptors are distributed in strategic areas belonging to the mesolimbic DA system such as the ventral striatum, midbrain, and pallidum, which have been associated with behaviors controlled by the presentation of drug-associated cues. Second, repeated exposure to drugs of abuse has been shown to produce neuroadaptations in the DA D(3) system. Third, the synthesis and characterization of highly potent and selective DA D(3) receptor antagonists has permitted to further define the role of the DA D(3) receptor in drug addiction. Provided that the available preclinical and preliminary clinical evidence can be translated into clinical proof of concept in human, selective DA D(3) receptor antagonists show promise for the treatment of substance use disorders as reflected by their potential to (1) regulate the motivation to self-administered drugs under schedules of reinforcement that require an increase in work demand and (2) disrupt the responsiveness to drug-associated stimuli that play a key role in the reinstatement of drug-seeking behavior triggered by re-exposure to the drug itself, re-exposure to environmental cues that had been previously associated with drug-taking behavior, or stress.
Collapse
Affiliation(s)
- Christian Heidbreder
- Reckitt Benckiser Pharmaceuticals-Global Research and Development, 10710 Midlothian Turnpike Suite 430, Richmond, VA 23235, USA.
| |
Collapse
|
28
|
Voxel scale complex networks of functional connectivity in the rat brain: neurochemical state dependence of global and local topological properties. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2012; 2012:615709. [PMID: 22919431 PMCID: PMC3415145 DOI: 10.1155/2012/615709] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Revised: 05/21/2012] [Accepted: 05/25/2012] [Indexed: 11/18/2022]
Abstract
Network analysis of functional imaging data reveals emergent features of the brain as a function of its topological properties. However, the brain is not a homogeneous network, and the dependence of functional connectivity parameters on neuroanatomical substrate and parcellation scale is a key issue. Moreover, the extent to which these topological properties depend on underlying neurochemical changes remains unclear. In the present study, we investigated both global statistical properties and the local, voxel-scale distribution of connectivity parameters of the rat brain. Different neurotransmitter systems were stimulated by pharmacological challenge (d-amphetamine, fluoxetine, and nicotine) to discriminate between stimulus-specific functional connectivity and more general features of the rat brain architecture. Although global connectivity parameters were similar, mapping of local connectivity parameters at high spatial resolution revealed strong neuroanatomical dependence of functional connectivity in the rat brain, with clear differentiation between the neocortex and older brain regions. Localized foci of high functional connectivity independent of drug challenge were found in the sensorimotor cortices, consistent with the high neuronal connectivity in these regions. Conversely, the topological properties and node roles in subcortical regions varied with neurochemical state and were dependent on the specific dynamics of the different functional processes elicited.
Collapse
|
29
|
Perles-Barbacaru TA, Procissi D, Demyanenko AV, Jacobs RE. Quantitative pharmacologic MRI in mice. NMR IN BIOMEDICINE 2012; 25:498-505. [PMID: 21793079 PMCID: PMC3292675 DOI: 10.1002/nbm.1760] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 05/03/2011] [Accepted: 05/19/2011] [Indexed: 05/31/2023]
Abstract
Pharmacologic MRI (phMRI) uses functional MRI techniques to provide a noninvasive in vivo measurement of the hemodynamic effects of drugs. The cerebral blood volume change (ΔCBV) serves as a surrogate for neuronal activity via neurovascular coupling mechanisms. By assessing the location and time course of brain activity in mouse mutant studies, phMRI can provide valuable insights into how different behavioral phenotypes are expressed in deferring brain activity response to drug challenge. In this report, we evaluate the utility of three different intravascular ultrasmall superparamagnetic iron oxide (USPIO) contrast agents for phMRI using a gradient-echo technique, with temporal resolution of one min at high magnetic field. The tissue half-life of the USPIOs was studied using a nonlinear detrending model. The three USPIOs are candidates for CBV weighted phMRI experiments, with r(2)/r(1) ratios ≥ 20 and apparent half-lives ≥ 1.5 h at the described doses. An echo-time of about 10 ms or longer results in a functional contrast to noise ratio (fCNR) > 75 after USPIO injection, with negligible decrease between 1.5-2 h. phMRI experiments were conducted at 7 T using cocaine as a psychotropic substance and acetazolamide, a global vasodilator, as a positive control. Cocaine acts as a dopamine-serotonin-norepinephrine reuptake inhibitor, increasing extracellular concentrations of these neurotransmitters, and thus increasing dopaminergic, serotonergic and noradrenergic neurotransmission. phMRI results showed that CBV was reduced in the normal mouse brain after cocaine challenge, with the largest effects in the nucleus accumbens, whereas after acetazolamide, blood volume was increased in both cerebral and extracerebral tissue.
Collapse
Affiliation(s)
| | - Daniel Procissi
- Biological Imaging Center, Beckman Institute, California Institute of Technology, Pasadena, CA 91125
| | - Andrey V. Demyanenko
- Biological Imaging Center, Beckman Institute, California Institute of Technology, Pasadena, CA 91125
| | - Russell E. Jacobs
- Biological Imaging Center, Beckman Institute, California Institute of Technology, Pasadena, CA 91125
| |
Collapse
|
30
|
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: 7.6] [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
|
31
|
Abstract
Neuroimaging allows researchers and clinicians to noninvasively assess structure and function of the brain. With the advances of imaging modalities such as magnetic resonance, nuclear, and optical imaging; the design of target-specific probes; and/or the introduction of reporter gene assays, these technologies are now capable of visualizing cellular and molecular processes in vivo. Undoubtedly, the system biological character of molecular neuroimaging, which allows for the study of molecular events in the intact organism, will enhance our understanding of physiology and pathophysiology of the brain and improve our ability to diagnose and treat diseases more specifically. Technical/scientific challenges to be faced are the development of highly sensitive imaging modalities, the design of specific imaging probe molecules capable of penetrating the CNS and reporting on endogenous cellular and molecular processes, and the development of tools for extracting quantitative, biologically relevant information from imaging data. Today, molecular neuroimaging is still an experimental approach with limited clinical impact; this is expected to change within the next decade. This article provides an overview of molecular neuroimaging approaches with a focus on rodent studies documenting the exploratory state of the field. Concepts are illustrated by discussing applications related to the pathophysiology of Alzheimer's disease.
Collapse
Affiliation(s)
- Jan Klohs
- Institute for Biomedical Engineering, ETH & University of Zürich, Switzerland
| | | |
Collapse
|
32
|
Baker S, Chin CL, Basso AM, Fox GB, Marek GJ, Day M. Xanomeline modulation of the blood oxygenation level-dependent signal in awake rats: development of pharmacological magnetic resonance imaging as a translatable pharmacodynamic biomarker for central activity and dose selection. J Pharmacol Exp Ther 2012; 341:263-73. [PMID: 22267203 DOI: 10.1124/jpet.111.188797] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In vivo translational imaging techniques, such as positron emission tomography and single-photon emission-computed tomography, are the only ways to adequately determine that a drug engages its target. Unfortunately, there are far more experimental mechanisms being tested in the clinic than there are radioligands, impeding the use of this risk-mitigating approach in modern drug discovery and development. Pharmacological magnetic resonance imaging (phMRI) offers an approach for developing new biomarkers with the potential to determine central activity and dose selection in animals and humans. Using phMRI, we characterized the effects of xanomeline on ketamine-induced activation on blood oxygen level-dependent (BOLD) signal. In the present studies, xanomeline alone dose-dependently increased the BOLD signal across several regions of interest, including association and motor and sensory cortical regions. It is noteworthy that xanomeline dose-dependently attenuated ketamine-induced brain activation patterns, effects that were antagonized by atropine. In conclusion, the muscarinic 1/4-preferring receptor agonist xanomeline suppressed the effects of the N-methyl-D-aspartate channel blocker ketamine in a number of brain regions, including the association cortex, motor cortex, and primary sensory cortices. The region-specific brain activation observed in this ketamine challenge phMRI study may provide a method of confirming central activity and dose selection for novel antipsychotic drugs in early clinical trials for schizophrenia, if the data obtained in animals can be recapitulated in humans.
Collapse
Affiliation(s)
- Scott Baker
- Translational Sciences, Advanced Technology, Global Pharmaceutical Research and Development, Abbott Laboratories, Abbott Park, IL, USA
| | | | | | | | | | | |
Collapse
|
33
|
Wang Z, Pang RD, Hernandez M, Ocampo MA, Holschneider DP. Anxiolytic-like effect of pregabalin on unconditioned fear in the rat: an autoradiographic brain perfusion mapping and functional connectivity study. Neuroimage 2011; 59:4168-88. [PMID: 22155030 DOI: 10.1016/j.neuroimage.2011.11.047] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2011] [Revised: 11/10/2011] [Accepted: 11/16/2011] [Indexed: 12/15/2022] Open
Abstract
Clinical and preclinical evidence suggests anxiolytic-like efficacy of pregabalin (PGB, Lyrica). However, its mechanism of action remains under investigation. The current study applied [(14)C]-iodoantipyrine cerebral blood flow (CBF) mapping to examine the effect of PGB on neural substrates underlying unconditioned fear in a rat model of footshock-induced fear. Regional CBF (rCBF) was analyzed by statistical parametric mapping. Functional connectivity and graph theoretical analysis were used to investigate how footshock and PGB affect brain activation at the network level. Pregabalin significantly attenuated footshock-induced ultrasonic vocalization, but showed no significant effect on freezing behavior. Footshock compared to no-shock controls elicited significant increases in rCBF in limbic/paralimbic regions implicated in the processing of unconditioned fear and ultrasonic vocalization, including the amygdala, hypothalamus, lateral septum, dorsal periaqueductal gray, the anterior insular (aINS) and medial prefrontal cortex (mPFC). The activation pattern was similar in vehicle- and PGB-treated subjects, with PGB significantly attenuating activation in the amygdala, hypothalamus, and aINS. The vehicle/no-shock group showed strong, positive intra-structural correlations within the cortex, hypothalamus, amygdala, thalamus, and brainstem. The cortex was negatively correlated with the hypothalamus and brainstem. Footshock reduced the total number of significant correlations, but induced greater intra-cortical connectivity of the aINS and mPFC, and new positive correlations between the hypothalamus and amygdala. In no-shock controls, PGB significantly reduced the positive intra-structural correlations within the cortex and amygdala, as well as the negative cortico-subcortical correlations. Following footshocks, PGB disrupted both the network recruitment of aINS and mPFC, and the positive hypothalamic-amygdaloid correlations. Our findings suggest that PGB may exert anxiolytic effect by attenuating cortico-cortical and cortico-subcortical communication and inhibiting network recruitment of the aINS, mPFC, amygdala, and hypothalamus following a fear-inducing stimulus. Functional brain mapping in rodents may provide new endpoints for preclinical evaluation of anxiolytic drug candidates with potentially improved translational power compared to behavioral measurements alone.
Collapse
Affiliation(s)
- Zhuo Wang
- Department of Psychiatry and Behavioral Sciences, University of Southern California, Los Angeles, CA, USA
| | | | | | | | | |
Collapse
|
34
|
Borsook D, Hargreaves R, Becerra L. Can Functional Magnetic Resonance Imaging Improve Success Rates in CNS Drug Discovery? Expert Opin Drug Discov 2011; 6:597-617. [PMID: 21765857 DOI: 10.1517/17460441.2011.584529] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION: The bar for developing new treatments for CNS disease is getting progressively higher and fewer novel mechanisms are being discovered, validated and developed. The high costs of drug discovery necessitate early decisions to ensure the best molecules and hypotheses are tested in expensive late stage clinical trials. The discovery of brain imaging biomarkers that can bridge preclinical to clinical CNS drug discovery and provide a 'language of translation' affords the opportunity to improve the objectivity of decision-making. AREAS COVERED: This review discusses the benefits, challenges and potential issues of using a science based biomarker strategy to change the paradigm of CNS drug development and increase success rates in the discovery of new medicines. The authors have summarized PubMed and Google Scholar based publication searches to identify recent advances in functional, structural and chemical brain imaging and have discussed how these techniques may be useful in defining CNS disease state and drug effects during drug development. EXPERT OPINION: The use of novel brain imaging biomarkers holds the bold promise of making neuroscience drug discovery smarter by increasing the objectivity of decision making thereby improving the probability of success of identifying useful drugs to treat CNS diseases. Functional imaging holds the promise to: (1) define pharmacodynamic markers as an index of target engagement (2) improve translational medicine paradigms to predict efficacy; (3) evaluate CNS efficacy and safety based on brain activation; (4) determine brain activity drug dose-response relationships and (5) provide an objective evaluation of symptom response and disease modification.
Collapse
Affiliation(s)
- David Borsook
- Center for Pain and the Brain, MGH, McLean and Children's Hospitals, Harvard Medical School And Merck Research Laboratories
| | | | | |
Collapse
|
35
|
Micheli F. Recent Advances in the Development of Dopamine D3 Receptor Antagonists: a Medicinal Chemistry Perspective. ChemMedChem 2011; 6:1152-62. [DOI: 10.1002/cmdc.201000538] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Indexed: 11/08/2022]
|
36
|
Abstract
Drug addiction is a syndrome of impaired response inhibition and salience attribution, which involves a complex neurocircuitry underlying drug reinforcement, drug craving, and compulsive drug-seeking and drug-taking behaviors despite adverse consequences. The concept of disease stages with transitions from acute rewarding effects to early- and end-stage addiction has had an important impact on the design of nonclinical animal models. This chapter reviews the main advances in nonclinical paradigms that aim to at model (1) positive and negative reinforcing effects of addictive drugs; (2) relapse to drug-seeking behavior; (3) reconsolidation of drug cue memories, and (4) compulsive/impulsive drug intake. In addition, recent small animal neuroimaging studies and invertebrate models will be briefly discussed (see also Bifone and Gozzi, Animal models of ADHD, 2011). Continuous improvement in modeling drug intake, craving, withdrawal symptoms, relapse, and comorbid psychiatric associations is a necessary step to better understand the etiology of the disease and to ultimately foster the discovery, validation and optimization of new efficacious pharmacotherapeutic approaches. The modeling of specific subprocesses or constructs that address clinically defined criteria will ultimately increase our understanding of the disease as a whole. Future research will have to address the questions of whether some of these constructs can be reliably used as outcome measures to assess the effects of a treatment in clinical settings, whether changes in those measures can be a target of therapeutic efforts, and whether they relate to biological markers of traits such as impulsivity, which contribute to increased drug-seeking and may predict binge-like patterns of drug intake.
Collapse
Affiliation(s)
- Christian Heidbreder
- Reckitt Benckiser Pharmaceuticals Inc., 10710 Midlothian Turnpike, Suite 430, Richmond, VA, 23235, USA,
| |
Collapse
|
37
|
Bifone A, Gozzi A. Functional and pharmacological MRI in understanding brain function at a systems level. Curr Top Behav Neurosci 2011; 7:323-57. [PMID: 21225416 DOI: 10.1007/7854_2010_103] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Functional magnetic resonance imaging (fMRI) methods have been extensively applied to study the human brain and its functional organization in healthy and disease states. A strong rationale exists for the extension of this approach to animal models as a translational tool to bridge clinical and preclinical research. Specifically, the development of pharmacological MRI (phMRI), i.e., the use of fMRI to map spatiotemporal patterns of brain activity induced by pharmacological agents, has provided a robust and flexible tool to resolve brain circuits and mechanism-specific functional changes produced by selective intervention in different neurotransmitter systems in vivo. This chapter describes the methodological aspects of fMRI and phMRI in preclinical species, and some of the key findings, with a special emphasis on the translational potential of these methods in neuropharmacological research.
Collapse
Affiliation(s)
- Angelo Bifone
- Center for Nanotechnology Innovation, Italian Institute of Technology, IIT@NEST, Piazza San Silvestro, 12, Pisa, 56127, Italy,
| | | |
Collapse
|
38
|
Perles-Barbacaru TA, Procissi D, Demyanenko AV, Hall FS, Uhl GR, Jacobs RE. Quantitative pharmacologic MRI: mapping the cerebral blood volume response to cocaine in dopamine transporter knockout mice. Neuroimage 2010; 55:622-8. [PMID: 21185387 DOI: 10.1016/j.neuroimage.2010.12.048] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Revised: 12/15/2010] [Accepted: 12/17/2010] [Indexed: 12/31/2022] Open
Abstract
The use of pharmacologic MRI (phMRI) in mouse models of brain disorders allows noninvasive in vivo assessment of drug-modulated local cerebral blood volume changes (ΔCBV) as one correlate of neuronal and neurovascular activities. In this report, we employed CBV-weighted phMRI to compare cocaine-modulated neuronal activity in dopamine transporter (DAT) knockout (KO) and wild-type mice. Cocaine acts to block the dopamine, norepinephrine, and serotonin transporters (DAT, NET, and SERT) that clear their respective neurotransmitters from the synapses, helping to terminate cognate neurotransmission. Cocaine consistently reduced CBV, with a similar pattern of regional ΔCBV in brain structures involved in mediating reward in both DAT genotypes. The largest effects (-20% to -30% ΔCBV) were seen in the nucleus accumbens and several cortical regions. Decreasing response amplitudes to cocaine were noted in more posterior components of the cortico-mesolimbic circuit. DAT KO mice had significantly attenuated ΔCBV amplitudes, shortened times to peak response, and reduced response duration in most regions. This study demonstrates that DAT knockout does not abolish the phMRI responses to cocaine, suggesting that adaptations to loss of DAT and/or retained cocaine activity in other monoamine neurotransmitter systems underlie these responses in DAT KO mice.
Collapse
|
39
|
Choi JK, Mandeville JB, Chen YI, Grundt P, Sarkar SK, Newman AH, Jenkins BG. Imaging brain regional and cortical laminar effects of selective D3 agonists and antagonists. Psychopharmacology (Berl) 2010; 212:59-72. [PMID: 20628733 PMCID: PMC3822611 DOI: 10.1007/s00213-010-1924-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Accepted: 06/15/2010] [Indexed: 02/04/2023]
Abstract
RATIONALE Dopamine D3 receptors (D3R) may be important therapeutic targets for both drug abuse and dyskinesias in Parkinson's disease; however, little is known about their functional circuitry. OBJECTIVES We wished to determine if D3R antagonists SB-277011 and PG-01037 and D3R-preferring agonist 7-OH-DPAT are D3R selective in vivo. We further wished to characterize the response to D3R drugs using whole brain imaging to identify novel D3R circuitry. METHODS We investigated D3R circuitry in rats using pharmacologic MRI and challenge with selective D3R antagonists and agonist at various doses to examine regional changes in cerebral blood volume (CBV). We compared regional activation patterns with D2R/D3R agonists, as well as with prior studies of mRNA expression and autoradiography. RESULTS D3R antagonists induced positive CBV changes and D3R agonist negative CBV changes in brain regions including nucleus accumbens, infralimbic cortex, thalamus, interpeduncular region, hypothalamus, and hippocampus (strongest in subiculum). All D3R-preferring drugs showed markedly greater responses in nucleus accumbens than in caudate/putamen consistent with D3R selectivity and contrary to what was observed with D2R agonists. At high doses of D3R agonist, functional changes were differentiated across cortical laminae, with layer V-VI yielding positive CBV changes and layer IV yielding negative CBV changes. These results are not inconsistent with differential D1R and D3R innervation in these layers respectively showed previously using post-mortem techniques. CONCLUSIONS MRI provides a new tool for testing the in vivo selectivity of novel D3R dopaminergic ligands where radiolabels may not be available. Further, the functional D3R circuitry strongly involves hypothalamus and subiculum as well as the limbic striatum.
Collapse
Affiliation(s)
- Ji-Kyung Choi
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Charlestown, 02129, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Bifone A, Gozzi A, Schwarz AJ. Functional connectivity in the rat brain: a complex network approach. Magn Reson Imaging 2010; 28:1200-9. [PMID: 20813478 DOI: 10.1016/j.mri.2010.07.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Revised: 06/30/2010] [Accepted: 07/13/2010] [Indexed: 02/04/2023]
Abstract
Functional connectivity analyses of fMRI data can provide a wealth of information on the brain functional organization and have been widely applied to the study of the human brain. More recently, these methods have been extended to preclinical species, thus providing a powerful translational tool. Here, we review methods and findings of functional connectivity studies in the rat. More specifically, we focus on correlation analysis of pharmacological MRI (phMRI) responses, an approach that has enabled mapping the patterns of connectivity underlying major neurotransmitter systems in vivo. We also review the use of novel statistical approaches based on a network representation of the functional connectivity and their application to the study of the rat brain functional architecture.
Collapse
Affiliation(s)
- Angelo Bifone
- Italian Institute of Technology, Center for Nanotechnology Innovation, IIT@NEST, Piazza San Silvestro 12, Pisa, Italy.
| | | | | |
Collapse
|
41
|
Murnane KS, Howell LL. Development of an apparatus and methodology for conducting functional magnetic resonance imaging (fMRI) with pharmacological stimuli in conscious rhesus monkeys. J Neurosci Methods 2010; 191:11-20. [PMID: 20566353 PMCID: PMC2915932 DOI: 10.1016/j.jneumeth.2010.06.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Revised: 04/28/2010] [Accepted: 06/01/2010] [Indexed: 11/23/2022]
Abstract
Functional magnetic resonance imaging (fMRI) is a technique with significant potential to advance our understanding of multiple brain systems. However, when human subjects undergo fMRI studies they are typically conscious whereas pre-clinical fMRI studies typically utilize anesthesia, which complicates comparisons across studies. Therefore, we have developed an apparatus suitable for imaging conscious rhesus monkeys. In order to minimize subject stress and spatial motion, each subject was acclimated to the necessary procedures over several months. The effectiveness of this process was then evaluated, in fully trained subjects, by quantifying objective physiological measures. These physiological metrics were stable both within and across sessions and did not differ from when these same subjects were immobilized using standard primate handling procedures. Subject motion and blood oxygenation level dependent (BOLD) fMRI measurements were then evaluated by scanning subjects under three different conditions: the absence of stimulation, presentation of a visual stimulus, or administration of intravenous (i.v.) cocaine (0.3mg/kg). Spatial motion differed neither by condition nor along the three principal axes. In addition, maximum translational and rotational motion never exceeded one half of the voxel size (0.75 mm) or 1.5 degrees, respectively. Furthermore, the localization of changes in blood oxygenation closely matched those reported in previous studies using similar stimuli. These findings document the feasibility of fMRI data collection in conscious rhesus monkeys using these procedures and allow for the further study of the neural effects of psychoactive drugs.
Collapse
Affiliation(s)
- Kevin Sean Murnane
- Division of Neuroscience, Yerkes National Primate Research Center, Atlanta, GA USA
| | - Leonard Lee Howell
- Division of Neuroscience, Yerkes National Primate Research Center, Atlanta, GA USA
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
| |
Collapse
|
42
|
Mason CW, Hassan HE, Kim KP, Cao J, Eddington ND, Newman AH, Voulalas PJ. Characterization of the transport, metabolism, and pharmacokinetics of the dopamine D3 receptor-selective fluorenyl- and 2-pyridylphenyl amides developed for treatment of psychostimulant abuse. J Pharmacol Exp Ther 2010; 333:854-64. [PMID: 20228156 PMCID: PMC2879935 DOI: 10.1124/jpet.109.165084] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Accepted: 03/11/2010] [Indexed: 11/22/2022] Open
Abstract
The recent discovery of novel high-affinity and selective dopamine D3 receptor (DA D3R) antagonists and partial agonists has provided tools with which to further elucidate the role DA D3R plays in substance abuse. The present study was conducted to evaluate the transport, metabolism, pharmacokinetics, and brain uptake of the DA D3R-selective fluorenyl amides, NGB 2904 [N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butyl)-9H-fluorene-2-carboxamide] fumarate) and JJC 4-077 [N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)-3-hydroxybutyl)-9H-fluorene-2-carboxamide hydrochloride], and the 2-pyridylphenyl amides, CJB 090 [N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butyl)-4-(pyridine-2-yl)benzamide hydrochloride] and PG 01037 [N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)-trans-but-2-enyl)-4-(pyridine-2-yl)benzamide hydrochloride], all of which have been studied in animal models of psychostimulant abuse. Additional screening with a panel of human and rat Supersomes was performed for NGB 2904 and PG 01037. Drug-stimulated ATPase activation assays and bidirectional transport and efflux assays were used to test for substrate specificity of NGB 2904 and PG 01037 for human and rat efflux transporters. All compounds exhibited moderate elimination half-lives, ranging from 1.49 to 3.27 h, and large volumes of distribution (5.95-14.19 l/kg). The brain-to-plasma ratios ranged from 2.93 to 11.81 and were higher than those previously reported for cocaine. Brain exposure levels of NGB 2904 and PG 01037 were significantly reduced after intraperitoneal administration compared with intravenous administration. The metabolism of these compounds was mediated primarily by CYP3A subfamilies. PG 01037 was a P-glycoprotein-transported substrate. Higher doses of these compounds are often required for in vivo action, suggesting decreased bioavailability via extravascular administration that may be attributed to high drug efflux and hepatic metabolism. These studies provide important preclinical information for optimization of next-generation D3R selective agents for the treatment of drug addiction.
Collapse
Affiliation(s)
- Clifford W Mason
- Pharmacokinetics-Biopharmaceutics Laboratory, Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Gozzi A, Crestan V, Turrini G, Clemens M, Bifone A. Antagonism at serotonin 5-HT(2A) receptors modulates functional activity of frontohippocampal circuit. Psychopharmacology (Berl) 2010; 209:37-50. [PMID: 20111859 DOI: 10.1007/s00213-009-1772-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Accepted: 12/20/2009] [Indexed: 12/22/2022]
Abstract
RATIONALE Several second-generation antipsychotics are characterised by a significant antagonistic effect at serotonin 5-HT(2A) receptors (5-HT(2A)R), a feature that has been associated with lower incidence of extra-pyramidal symptoms and a putative amelioration of positive and negative symptoms experienced by schizophrenic patients. However, the neurofunctional substrate of 5-HT(2A) antagonism and its exact contribution to the complex pharmacological profile of these drugs remain to be elucidated. OBJECTIVES Here, we used pharmacological magnetic resonance imaging to map the modulatory effects of the selective 5-HT(2A)R antagonist Ml00907 on the spatiotemporal patterns of brain activity elicited by acute phencyclidine (PCP) challenge in the rat. PCP is a non-competitive NMDA receptor antagonist that induces dysregulation of corticolimbic glutamatergic neurotransmission and produces cognitive impairment and psychotic-like symptoms reminiscent of those observed in schizophrenia. RESULTS Pre-administration of M100907 produced focal and region-dependent attenuation of PCP-induced response in frontoseptohippocampal areas. As early studies highlighted a permissive role of 5-HT(2A)R on frontal dopamine release, the role of post-synaptic dopamine D(1) receptors on PCP-induced response was examined by using the potent antagonist SCH23390. Interestingly, SCH23390 did not affect PCP's response in any of the regions examined. This finding rules out a significant contribution of dopamine in the functional changes mapped and, indirectly, the inhibitory effect of M100907, in favour of a glutamatergic origin. CONCLUSIONS Our data expand recent evidence suggesting a key role of 5-HT(2A)R in modulating glutamate-mediated cognitive performance in the prefrontal cortex and highlight the whole frontoseptohippocampal circuit as a key functional substrate of 5-HT(2A)R antagonism in normal and disease states.
Collapse
Affiliation(s)
- Alessandro Gozzi
- Biology, Neurosciences CEDD, GlaxoSmithKline Medicines Research Centre, Verona, Italy
| | | | | | | | | |
Collapse
|
44
|
Heidbreder CA, Newman AH. Current perspectives on selective dopamine D(3) receptor antagonists as pharmacotherapeutics for addictions and related disorders. Ann N Y Acad Sci 2010; 1187:4-34. [PMID: 20201845 PMCID: PMC3148950 DOI: 10.1111/j.1749-6632.2009.05149.x] [Citation(s) in RCA: 233] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Repeated exposure to drugs of abuse produces long-term molecular and neurochemical changes that may explain the core features of addiction, such as the compulsive seeking and taking of the drug, as well as the risk of relapse. A growing number of new molecular and cellular targets of addictive drugs have been identified, and rapid advances are being made in relating those targets to specific behavioral phenotypes in animal models of addiction. In this context, the pattern of expression of the dopamine (DA) D(3) receptor in the rodent and human brain and changes in this pattern in response to drugs of abuse have contributed primarily to direct research efforts toward the development of selective DA D(3) receptor antagonists. Growing preclinical evidence indicates that these compounds may actually regulate the motivation to self-administer drugs and disrupt drug-associated cue-induced craving. This report will be divided into three parts. First, preclinical evidence in support of the efficacy of selective DA D(3) receptor antagonists in animal models of drug addiction will be reviewed. The effects of mixed DA D(2)/D(3) receptor antagonists will not be discussed here because most of these compounds have low selectivity at the D(3) versus D(2) receptor, and their efficacy profile is related primarily to functional antagonism at D(2) receptors and possibly interactions with other neurotransmitter systems. Second, major advances in medicinal chemistry for the identification and optimization of selective DA D(3) receptor antagonists and partial agonists will be analyzed. Third, translational research from preclinical efficacy studies to so-called proof-of-concept studies for drug addiction indications will be discussed.
Collapse
Affiliation(s)
- Christian A Heidbreder
- Reckitt Benckiser Pharmaceuticals, Global Research & Development, Richmond, Virginia 23235, USA.
| | | |
Collapse
|
45
|
Diaconescu AO, Menon M, Jensen J, Kapur S, McIntosh AR. Dopamine-induced changes in neural network patterns supporting aversive conditioning. Brain Res 2009; 1313:143-61. [PMID: 19961836 DOI: 10.1016/j.brainres.2009.11.064] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Revised: 11/21/2009] [Accepted: 11/24/2009] [Indexed: 11/30/2022]
Abstract
The aim of the present paper is to assess the effects of altered dopamine (DA) transmission on the functional connectivity among brain regions mediating aversive conditioning in humans. To this aim, we analyzed a previous published data set from a double-blind design combined with functional magnetic resonance imaging (fMRI) recordings in which healthy volunteers were randomly assigned to one of three drug groups: amphetamine (an indirect DA agonist), haloperidol (DA D2 receptor antagonist), and placebo. Participants were exposed to an aversive classical conditioning paradigm using cutaneous electrical stimulation as the unconditioned stimulus (US), and visual cues as the conditioned stimuli (CS) where one colour (CS+) was followed by the US in 33% of the trials and another colour (CS-) had no consequences. All participants reported awareness of stimulus contingencies. Group analysis of fMRI data revealed that the left ventral striatum (VS) and amygdala activated in response to the CS+ in all the three groups. Because of their activation patterns and documented involvement in aversive conditioning, both regions were used as seeds in the functional connectivity analysis. To constrain the functional networks obtained to relate to the conditioned response, we also correlated seed activity with the Galvanic Skin Response (GSR). In the placebo group, the right ventral tegmental area/substantia nigra (VTA/SN), bilateral caudate, right parahippocampal gyrus, left inferior parietal lobule (IPL), bilateral postcentral gyrus, bilateral middle frontal (BA 46), orbitofrontal, and ventromedial prefrontal cortices (PFC, BA 10/11) correlated with the VS and amygdala seeds in response to the CS+ compared to the CS-. Enhancing dopamine transmission via amphetamine was associated with reduced task differences and significant functional connectivity for both CS+ and CS- conditions between the left VS seed and regions modulated by DA, such as the left VTA/SN, right caudate, left amygdala, left middle frontal gyrus (BA 46), and bilateral ventromedial PFC (BA 10). Blocking dopamine transmission via haloperidol was associated with significant functional connectivity across an alternate network of regions including the left amygdala seed and the right insula, the left ACC (BA 24/32), bilateral IPL (BA 40), precuneus (BA 7), post-central gyrus, middle frontal gyrus (BA 46), and supplementary motor area (SMA, BA 6) to the CS+ versus the CS-. These data provide insight into the distinct effects of DA agents on the functional connectivity between striatal, limbic, and prefrontal areas.
Collapse
|
46
|
Schwarz AJ, Gozzi A, Bifone A. Community structure in networks of functional connectivity: Resolving functional organization in the rat brain with pharmacological MRI. Neuroimage 2009; 47:302-11. [DOI: 10.1016/j.neuroimage.2009.03.064] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2009] [Revised: 03/10/2009] [Accepted: 03/22/2009] [Indexed: 12/15/2022] Open
|
47
|
Spanagel R. Alcoholism: A Systems Approach From Molecular Physiology to Addictive Behavior. Physiol Rev 2009; 89:649-705. [DOI: 10.1152/physrev.00013.2008] [Citation(s) in RCA: 491] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Alcohol consumption is an integral part of daily life in many societies. The benefits associated with the production, sale, and use of alcoholic beverages come at an enormous cost to these societies. The World Health Organization ranks alcohol as one of the primary causes of the global burden of disease in industrialized countries. Alcohol-related diseases, especially alcoholism, are the result of cumulative responses to alcohol exposure, the genetic make-up of an individual, and the environmental perturbations over time. This complex gene × environment interaction, which has to be seen in a life-span perspective, leads to a large heterogeneity among alcohol-dependent patients, in terms of both the symptom dimensions and the severity of this disorder. Therefore, a reductionistic approach is not very practical if a better understanding of the pathological processes leading to an addictive behavior is to be achieved. Instead, a systems-oriented perspective in which the interactions and dynamics of all endogenous and environmental factors involved are centrally integrated, will lead to further progress in alcohol research. This review adheres to a systems biology perspective such that the interaction of alcohol with primary and secondary targets within the brain is described in relation to the behavioral consequences. As a result of the interaction of alcohol with these targets, alterations in gene expression and synaptic plasticity take place that lead to long-lasting alteration in neuronal network activity. As a subsequent consequence, alcohol-seeking responses ensue that can finally lead via complex environmental interactions to an addictive behavior.
Collapse
|
48
|
Complex brain networks: graph theoretical analysis of structural and functional systems. Nat Rev Neurosci 2009; 10:186-98. [PMID: 19190637 DOI: 10.1038/nrn2575] [Citation(s) in RCA: 6673] [Impact Index Per Article: 444.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Recent developments in the quantitative analysis of complex networks, based largely on graph theory, have been rapidly translated to studies of brain network organization. The brain's structural and functional systems have features of complex networks--such as small-world topology, highly connected hubs and modularity--both at the whole-brain scale of human neuroimaging and at a cellular scale in non-human animals. In this article, we review studies investigating complex brain networks in diverse experimental modalities (including structural and functional MRI, diffusion tensor imaging, magnetoencephalography and electroencephalography in humans) and provide an accessible introduction to the basic principles of graph theory. We also highlight some of the technical challenges and key questions to be addressed by future developments in this rapidly moving field.
Collapse
|
49
|
Congestri F, Formenti F, Sonntag V, Hdou G, Crespi F. Selective D3 Receptor Antagonist SB-277011-A Potentiates the Effect of Cocaine on Extracellular Dopamine in the Nucleus Accumbens: a Dual Core-Shell Voltammetry Study in Anesthetized Rats. SENSORS (BASEL, SWITZERLAND) 2008; 8:6936-6951. [PMID: 27873908 PMCID: PMC3787424 DOI: 10.3390/s8116936] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Revised: 10/08/2008] [Accepted: 11/03/2008] [Indexed: 11/16/2022]
Abstract
Dopamine (DA) D3 receptors have been associated with drug intake and abuse and selectively distribute in the brain circuits responding to drug administration. Here we examined the effects of an acute systemic administration of cocaine (15 mg/kg) alone or preceded by treatment with the selective D3 receptor antagonist SB-277011-A (10 mg/kg) on DA levels concurrently in the rat nucleus accumbens shell and core sub-regions (NAcshell and NAccore, respectively). It is shown that cocaine increases extracellular DA in both compartments and that blocking D3 receptors with SB-277011-A, although the latter is devoid of dopaminergic effects per se, potentiates these effects. No differences in the amplitude of the response were observed between NAcshell and NAccore compartments, though the dopaminergic response in the NAcshell was transient whereas that in the NAccore rose slowly to reach a plateau. These results demonstrate the feasibility to use multiprobe voltammetry to measure discrete monoaminergic responses in discrete areas of the brain and confirm the effect of D3 receptors antagonist at modifying the neurochemical effects of cocaine.
Collapse
Affiliation(s)
| | | | - Viviana Sonntag
- Biology Dept, GlaxoSmithKline, Medicines Research Centre, Verona, Italy
| | - Gael Hdou
- Biology Dept, GlaxoSmithKline, Medicines Research Centre, Verona, Italy
| | - Francesco Crespi
- Biology Dept, GlaxoSmithKline, Medicines Research Centre, Verona, Italy.
| |
Collapse
|
50
|
Pharmacological MRI in animal models: A useful tool for 5-HT research? Neuropharmacology 2008; 55:1038-47. [DOI: 10.1016/j.neuropharm.2008.08.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2008] [Revised: 08/06/2008] [Accepted: 08/07/2008] [Indexed: 01/19/2023]
|