1
|
Oliver B, Devitt C, Park G, Razak A, Liu SM, Bergese SD. Drugs in Development to Manage Acute Pain. Drugs 2024:10.1007/s40265-024-02118-0. [PMID: 39560856 DOI: 10.1007/s40265-024-02118-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2024] [Indexed: 11/20/2024]
Abstract
Acute pain, defined as short-term pain arising from injury or other noxious stimuli, affects patient outcomes, quality of life, and healthcare costs. Safe, effective treatment of acute pain is essential in preventing increased morbidity, mortality, and the transition to chronic pain. In this review, we explore some of the latest therapeutic agents, formulations, combinations, and administration routes of drugs emerging in clinical practice in the USA for the treatment of acute pain. These agents include VX-548 (Suzetrigine), Cebranopadol, AAT-076, Combogesic intravenous (IV), sublingual ketamine, XG004 (naproxen/pregabalin conjugate), and HTX-011 (Zynrelef). We analyze the pharmacodynamics, pharmacokinetics, development status, and clinical implications of these drugs, emphasizing the importance of finding an agent that provides both a strong safety profile and effective relief from acute pain. Our findings show promise but also highlight the need for further large-scale research to allow these drugs to be utilized in a clinical context for patients experiencing acute pain.
Collapse
Affiliation(s)
- Brian Oliver
- Department of Anesthesiology, Stony Brook Medicine, Stony Brook, NY, USA
| | - Catherine Devitt
- Department of Anesthesiology, Stony Brook Medicine, Stony Brook, NY, USA
| | - Grace Park
- Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Alina Razak
- Department of Anesthesiology, Stony Brook Medicine, Stony Brook, NY, USA
| | - Sun Mei Liu
- Department of Anesthesiology, Stony Brook Medicine, Stony Brook, NY, USA
| | - Sergio D Bergese
- Department of Anesthesiology, Stony Brook Medicine, Stony Brook, NY, USA.
- Anesthesiology and Neurological Surgery, Stony Brook University Health Science Center, Stony Brook, NY, 11794-8480, USA.
| |
Collapse
|
2
|
Gärtner M, Weigand A, Meiering MS, Weigner D, Carstens L, Keicher C, Hertrampf R, Beckmann C, Mennes M, Wunder A, Grimm S. Negative emotionality shapes the modulatory effects of ketamine and lamotrigine in subregions of the anterior cingulate cortex. Transl Psychiatry 2024; 14:258. [PMID: 38890270 PMCID: PMC11189565 DOI: 10.1038/s41398-024-02977-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 05/31/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024] Open
Abstract
Neuroimaging studies have identified the anterior cingulate cortex (ACC) as one of the major targets of ketamine in the human brain, which may be related to ketamine's antidepressant (AD) mechanisms of action. However, due to different methodological approaches, different investigated populations, and varying measurement timepoints, results are not consistent, and the functional significance of the observed brain changes remains a matter of open debate. Inhibition of glutamate release during acute ketamine administration by lamotrigine provides the opportunity to gain additional insight into the functional significance of ketamine-induced brain changes. Furthermore, the assessment of trait negative emotionality holds promise to link findings in healthy participants to potential AD mechanisms of ketamine. In this double-blind, placebo-controlled, randomized, single dose, parallel-group study, we collected resting-state fMRI data before, during, and 24 h after ketamine administration in a sample of 75 healthy male and female participants who were randomly allocated to one of three treatment conditions (ketamine, ketamine with lamotrigine pre- treatment, placebo). Spontaneous brain activity was extracted from two ventral and one dorsal subregions of the ACC. Our results showed activity decreases during the administration of ketamine in all three ACC subregions. However, only in the ventral subregions of the ACC this effect was attenuated by lamotrigine. 24 h after administration, ACC activity returned to baseline levels, but group differences were observed between the lamotrigine and the ketamine group. Trait negative emotionality was closely linked to activity changes in the subgenual ACC after ketamine administration. These results contribute to an understanding of the functional significance of ketamine effects in different subregions of the ACC by combining an approach to modulate glutamate release with the assessment of multiple timepoints and associations with trait negative emotionality in healthy participants.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Andreas Wunder
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Simone Grimm
- Medical School Berlin, Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland
| |
Collapse
|
3
|
Ait Bentaleb K, Boisvert M, Tourjman V, Potvin S. A Meta-Analysis of Functional Neuroimaging Studies of Ketamine Administration in Healthy Volunteers. J Psychoactive Drugs 2024; 56:211-224. [PMID: 36921026 DOI: 10.1080/02791072.2023.2190758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/22/2023] [Indexed: 03/17/2023]
Abstract
Ketamine administration leads to a psychotomimetic state when taken in large bolus doses, making it a valid model of psychosis. Therefore, understanding ketamine's effects on brain functioning is particularly relevant. This meta-analysis focused on neuroimaging studies that examined ketamine-induced brain activation at rest and during a task. Included are 10 resting-state studies and 23 task-based studies, 9 of which were measuring executive functions. Using a stringent statistical threshold (TFCE <0.05), the results showed increased activity at rest in the dorsal anterior cingulate cortex (ACC), and increased activation of the right Heschl's gyrus during executive tasks, following ketamine administration. Uncorrected results showed increased activation at rest in the right (anterior) insula and the right-fusiform gyrus, as well as increased activation during executive tasks in the rostral ACC. Rest-state studies highlighted alterations in core hubs of the salience network, while task-based studies suggested an impact on task-irrelevant brain regions. Increased activation in the rostral ACC may indicate a failure to deactivate the default mode network during executive tasks following ketamine administration. The results are coherent with alterations found in schizophrenia, which confer external validity to the ketamine model of psychosis. Studies investigating the neural mechanisms of ketamine's antidepressant action are warranted.
Collapse
Affiliation(s)
- Karim Ait Bentaleb
- Centre de recherche de l'Institut Universitaire en Santé Mentale de Montréal, Montréal, Canada
- Department of psychiatry and addiction, Université de Montréal, Montréal, Canada
| | - Mélanie Boisvert
- Centre de recherche de l'Institut Universitaire en Santé Mentale de Montréal, Montréal, Canada
- Department of psychiatry and addiction, Université de Montréal, Montréal, Canada
| | - Valérie Tourjman
- Centre de recherche de l'Institut Universitaire en Santé Mentale de Montréal, Montréal, Canada
- Department of psychiatry and addiction, Université de Montréal, Montréal, Canada
| | - Stéphane Potvin
- Centre de recherche de l'Institut Universitaire en Santé Mentale de Montréal, Montréal, Canada
- Department of psychiatry and addiction, Université de Montréal, Montréal, Canada
| |
Collapse
|
4
|
Gärtner M, Weigand A, Meiering MS, Weigner D, Carstens L, Keicher C, Hertrampf R, Beckmann C, Mennes M, Wunder A, Grimm S. Region- and time- specific effects of ketamine on cerebral blood flow: a randomized controlled trial. Neuropsychopharmacology 2023; 48:1735-1741. [PMID: 37231079 PMCID: PMC10579356 DOI: 10.1038/s41386-023-01605-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 05/04/2023] [Accepted: 05/07/2023] [Indexed: 05/27/2023]
Abstract
There is intriguing evidence suggesting that ketamine might have distinct acute and delayed neurofunctional effects, as its acute administration transiently induces schizophrenia-like symptoms, while antidepressant effects slowly emerge and are most pronounced 24 h after administration. Studies attempting to characterize ketamine's mechanism of action by using blood oxygen level dependent (BOLD) imaging have yielded inconsistent results regarding implicated brain regions and direction of effects. This may be due to intrinsic properties of the BOLD contrast, while cerebral blood flow (CBF), as measured with arterial spin labeling, is a single physiological marker more directly related to neural activity. As effects of acute ketamine challenge are sensitive to modulation by pretreatment with lamotrigine, which inhibits glutamate release, a combination of these approaches should be particularly suited to offer novel insights. In total, 75 healthy participants were investigated in a double blind, placebo-controlled, randomized, parallel-group study and underwent two scanning sessions (acute/post 24 h.). Acute ketamine administration was associated with higher perfusion in interior frontal gyrus (IFG) and dorsolateral prefrontal cortex (DLPFC), but no other investigated brain region. Inhibition of glutamate release by pretreatment with lamotrigine abolished ketamine's effect on perfusion. At the delayed time point, pretreatment with lamotrigine was associated with lower perfusion in IFG. These findings underscore the idea that regionally selective patterns of CBF changes reflect proximate effects of modulated glutamate release on neuronal activity. Furthermore, region- specific sustained effects indicate both a swift restoration of disturbed homeostasis in DLPFC as well changes occurring beyond the immediate effects on glutamate signaling in IFG.
Collapse
Affiliation(s)
- Matti Gärtner
- Medical School Berlin, Berlin, Germany.
- Department of Psychiatry and Psychotherapy, Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany.
| | | | | | | | | | | | | | | | | | - Andreas Wunder
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Simone Grimm
- Medical School Berlin, Berlin, Germany
- Department of Psychiatry and Psychotherapy, Charité, Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital of Psychiatry, University of Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Linguiti S, Vogel JW, Sydnor VJ, Pines A, Wellman N, Basbaum A, Eickhoff CR, Eickhoff SB, Edwards RR, Larsen B, McKinstry-Wu A, Scott JC, Roalf DR, Sharma V, Strain EC, Corder G, Dworkin RH, Satterthwaite TD. Functional imaging studies of acute administration of classic psychedelics, ketamine, and MDMA: Methodological limitations and convergent results. Neurosci Biobehav Rev 2023; 154:105421. [PMID: 37802267 DOI: 10.1016/j.neubiorev.2023.105421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/13/2023] [Accepted: 10/02/2023] [Indexed: 10/08/2023]
Abstract
Functional magnetic resonance imaging (fMRI) is increasingly used to non-invasively study the acute impact of psychedelics on the human brain. While fMRI is a promising tool for measuring brain function in response to psychedelics, it also has known methodological challenges. We conducted a systematic review of fMRI studies examining acute responses to experimentally administered psychedelics in order to identify convergent findings and characterize heterogeneity in the literature. We reviewed 91 full-text papers; these studies were notable for substantial heterogeneity in design, task, dosage, drug timing, and statistical approach. Data recycling was common, with 51 unique samples across 91 studies. Fifty-seven studies (54%) did not meet contemporary standards for Type I error correction or control of motion artifact. Psilocybin and LSD were consistently reported to moderate the connectivity architecture of the sensorimotor-association cortical axis. Studies also consistently reported that ketamine administration increased activation in the dorsomedial prefrontal cortex. Moving forward, use of best practices such as pre-registration, standardized image processing and statistical testing, and data sharing will be important in this rapidly developing field.
Collapse
Affiliation(s)
- Sophia Linguiti
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA, United States; Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Jacob W Vogel
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA, United States; Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States; Department of Clinical Sciences, Malmö, SciLifeLab, Lund University, Lund, Sweden
| | - Valerie J Sydnor
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA, United States; Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Adam Pines
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA, United States; Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States; Department of Psychiatry, Stanford University, Stanford, CA, United States
| | - Nick Wellman
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA, United States; Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Allan Basbaum
- Department of Anatomy, University of California, San Francisco, United States
| | - Claudia R Eickhoff
- Institute of Neuroscience and Medicine, (INM-1, INM-7), Research Centre Jülich, Jülich, Germany; Institute of Clinical Neuroscience and Medical Psychology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Simon B Eickhoff
- Institute of Neuroscience and Medicine, (INM-1, INM-7), Research Centre Jülich, Jülich, Germany; Institute of Systems Neuroscience, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Robert R Edwards
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Bart Larsen
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA, United States; Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Andrew McKinstry-Wu
- Department of Anesthesiology and Critical Care, Neuroscience of Unconsciousness and Reanimation Research Alliance (NEURRAL), University of Pennsylvania, Philadelphia, United States
| | - J Cobb Scott
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States; VISN4 Mental Illness Research, Education, and Clinical Center at the Corporal Michael J. Crescenz VA (Veterans Affairs) Medical Center, Philadelphia, PA, United States
| | - David R Roalf
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Vaishnavi Sharma
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA, United States; Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Eric C Strain
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, 5510 Nathan Shock Drive, Baltimore, MD, United States
| | - Gregory Corder
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Robert H Dworkin
- Department of Anesthesiology and Perioperative Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, United States
| | - Theodore D Satterthwaite
- Penn Lifespan Informatics and Neuroimaging Center (PennLINC), Philadelphia, PA, United States; Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States.
| |
Collapse
|
6
|
Boucherie DE, Reneman L, Ruhé HG, Schrantee A. Neurometabolite changes in response to antidepressant medication: A systematic review of 1H-MRS findings. Neuroimage Clin 2023; 40:103517. [PMID: 37812859 PMCID: PMC10563053 DOI: 10.1016/j.nicl.2023.103517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/22/2023] [Accepted: 09/23/2023] [Indexed: 10/11/2023]
Abstract
Selective serotonin reuptake inhibitors (SSRIs), serotonin and noradrenaline reuptake inhibitors (SNRIs), and (es)ketamine are used to treat major depressive disorder (MDD). These different types of medication may involve common neural pathways related to glutamatergic and GABAergic neurotransmitter systems, both of which have been implicated in MDD pathology. We conducted a systematic review of pharmacological proton Magnetic Resonance Spectroscopy (1H-MRS) studies in healthy volunteers and individuals with MDD to explore the potential impact of these medications on glutamatergic and GABAergic systems. We searched PubMed, Web of Science and Embase and included randomized controlled trials or cohort studies, which assessed the effects of SSRIs, SNRIs, or (es)ketamine on glutamate, glutamine, Glx or GABA using single-voxel 1H-MRS or Magnetic Resonance Spectroscopic Imaging (MRSI). Additionally, studies were included when they used a field strength > 1.5 T, and when a comparison of metabolite levels between antidepressant treatment and placebo or baseline with post-medication metabolite levels was done. We excluded animal studies, duplicate publications, or articles with 1H-MRS data already described in another included article. Twenty-nine studies were included in this review. Fifteen studies investigated the effect of administration or treatment with SSRIs or SNRIs, and fourteen studies investigated the effect of (es)ketamine on glutamatergic and GABAergic metabolite levels. Studies on SSRIs and SNRIs were highly variable, generally underpowered, and yielded no consistent findings across brain regions or specific populations. Although studies on (es)ketamine were also highly variable, some demonstrated an increase in glutamate levels in the anterior cingulate cortex in a time-dependent manner after administration. Our findings highlight the need for standardized study and acquisition protocols. Additionally, measuring metabolites dynamically over time or combining 1H-MRS with whole brain functional imaging techniques could provide valuable insights into the effects of these medications on glutamate and GABAergic neurometabolism.
Collapse
Affiliation(s)
- Daphne E Boucherie
- Amsterdam UMC, Location AMC, Department of Radiology and Nuclear Medicine, Meibergdreef 9, 1109 AZ Amsterdam, the Netherlands.
| | - Liesbeth Reneman
- Department of Psychiatry, Radboudumc, Radboud University, Reinier Postlaan 4, 6525 GC Nijmegen, the Netherlands
| | - Henricus G Ruhé
- Amsterdam UMC, Location AMC, Department of Radiology and Nuclear Medicine, Meibergdreef 9, 1109 AZ Amsterdam, the Netherlands; Department of Psychiatry, Radboudumc, Radboud University, Reinier Postlaan 4, 6525 GC Nijmegen, the Netherlands; Donders Institute for Brain Cognition and Behaviour, Radboud University, Kapittelweg 29, 6525 EN Nijmegen, the Netherlands
| | - Anouk Schrantee
- Amsterdam UMC, Location AMC, Department of Radiology and Nuclear Medicine, Meibergdreef 9, 1109 AZ Amsterdam, the Netherlands
| |
Collapse
|
7
|
Kotoula V, Evans JW, Punturieri CE, Zarate CA. Review: The use of functional magnetic resonance imaging (fMRI) in clinical trials and experimental research studies for depression. FRONTIERS IN NEUROIMAGING 2023; 2:1110258. [PMID: 37554642 PMCID: PMC10406217 DOI: 10.3389/fnimg.2023.1110258] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 06/12/2023] [Indexed: 08/10/2023]
Abstract
Functional magnetic resonance imaging (fMRI) is a non-invasive technique that can be used to examine neural responses with and without the use of a functional task. Indeed, fMRI has been used in clinical trials and pharmacological research studies. In mental health, it has been used to identify brain areas linked to specific symptoms but also has the potential to help identify possible treatment targets. Despite fMRI's many advantages, such findings are rarely the primary outcome measure in clinical trials or research studies. This article reviews fMRI studies in depression that sought to assess the efficacy and mechanism of action of compounds with antidepressant effects. Our search results focused on selective serotonin reuptake inhibitors (SSRIs), the most commonly prescribed treatments for depression and ketamine, a fast-acting antidepressant treatment. Normalization of amygdala hyperactivity in response to negative emotional stimuli was found to underlie successful treatment response to SSRIs as well as ketamine, indicating a potential common pathway for both conventional and fast-acting antidepressants. Ketamine's rapid antidepressant effects make it a particularly useful compound for studying depression with fMRI; its effects on brain activity and connectivity trended toward normalizing the increases and decreases in brain activity and connectivity associated with depression. These findings highlight the considerable promise of fMRI as a tool for identifying treatment targets in depression. However, additional studies with improved methodology and study design are needed before fMRI findings can be translated into meaningful clinical trial outcomes.
Collapse
|
8
|
Ossandón JP, Stange L, Gudi-Mindermann H, Rimmele JM, Sourav S, Bottari D, Kekunnaya R, Röder B. The development of oscillatory and aperiodic resting state activity is linked to a sensitive period in humans. Neuroimage 2023; 275:120171. [PMID: 37196987 DOI: 10.1016/j.neuroimage.2023.120171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/27/2023] [Accepted: 05/15/2023] [Indexed: 05/19/2023] Open
Abstract
Congenital blindness leads to profound changes in electroencephalographic (EEG) resting state activity. A well-known consequence of congenital blindness in humans is the reduction of alpha activity which seems to go together with increased gamma activity during rest. These results have been interpreted as indicating a higher excitatory/inhibitory (E/I) ratio in visual cortex compared to normally sighted controls. Yet it is unknown whether the spectral profile of EEG during rest would recover if sight were restored. To test this question, the present study evaluated periodic and aperiodic components of the EEG resting state power spectrum. Previous research has linked the aperiodic components, which exhibit a power-law distribution and are operationalized as a linear fit of the spectrum in log-log space, to cortical E/I ratio. Moreover, by correcting for the aperiodic components from the power spectrum, a more valid estimate of the periodic activity is possible. Here we analyzed resting state EEG activity from two studies involving (1) 27 permanently congenitally blind adults (CB) and 27 age-matched normally sighted controls (MCB); (2) 38 individuals with reversed blindness due to bilateral, dense, congenital cataracts (CC) and 77 age-matched sighted controls (MCC). Based on a data driven approach, aperiodic components of the spectra were extracted for the low frequency (Lf-Slope 1.5 to 19.5 Hz) and high frequency (Hf-Slope 20 to 45 Hz) range. The Lf-Slope of the aperiodic component was significantly steeper (more negative slope), and the Hf-Slope of the aperiodic component was significantly flatter (less negative slope) in CB and CC participants compared to the typically sighted controls. Alpha power was significantly reduced, and gamma power was higher in the CB and the CC groups. These results suggest a sensitive period for the typical development of the spectral profile during rest and thus likely an irreversible change in the E/I ratio in visual cortex due to congenital blindness. We speculate that these changes are a consequence of impaired inhibitory circuits and imbalanced feedforward and feedback processing in early visual areas of individuals with a history of congenital blindness.
Collapse
Affiliation(s)
- José P Ossandón
- Biological Psychology and Neuropsychology, University of Hamburg, Hamburg, Germany.
| | - Liesa Stange
- Biological Psychology and Neuropsychology, University of Hamburg, Hamburg, Germany
| | - Helene Gudi-Mindermann
- Biological Psychology and Neuropsychology, University of Hamburg, Hamburg, Germany; Institute of Public Health and Nursing Research, University of Bremen, Bremen, Germany
| | - Johanna M Rimmele
- Department of Neuroscience, Max-Planck-Institute for Empirical Aesthetics, Frankfurt, Germany; Department of Neurophysiology and Pathophysiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Max Planck NYU Center for Language, Music, and Emotion Frankfurt am Main, Germany, New York, NY, USA
| | - Suddha Sourav
- Biological Psychology and Neuropsychology, University of Hamburg, Hamburg, Germany
| | - Davide Bottari
- Biological Psychology and Neuropsychology, University of Hamburg, Hamburg, Germany; IMT School for Advanced Studies Lucca, Italy
| | - Ramesh Kekunnaya
- Child Sight Institute, Jasti V Ramanamma Children's Eye Care Center, LV Prasad Eye Institute, Hyderabad, India
| | - Brigitte Röder
- Biological Psychology and Neuropsychology, University of Hamburg, Hamburg, Germany; Child Sight Institute, Jasti V Ramanamma Children's Eye Care Center, LV Prasad Eye Institute, Hyderabad, India
| |
Collapse
|
9
|
Gonzalez-Burgos I, Bainier M, Gross S, Schoenenberger P, Ochoa JA, Valencia M, Redondo RL. Glutamatergic and GABAergic Receptor Modulation Present Unique Electrophysiological Fingerprints in a Concentration-Dependent and Region-Specific Manner. eNeuro 2023; 10:ENEURO.0406-22.2023. [PMID: 36931729 PMCID: PMC10124153 DOI: 10.1523/eneuro.0406-22.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 02/13/2023] [Accepted: 02/17/2023] [Indexed: 03/19/2023] Open
Abstract
Brain function depends on complex circuit interactions between excitatory and inhibitory neurons embedded in local and long-range networks. Systemic GABAA-receptor (GABAAR) or NMDA-receptor (NMDAR) modulation alters the excitatory-inhibitory balance (EIB), measurable with electroencephalography (EEG). However, EEG signatures are complex in localization and spectral composition. We developed and applied analytical tools to investigate the effects of two EIB modulators, MK801 (NMDAR antagonist) and diazepam (GABAAR modulator), on periodic and aperiodic EEG features in freely-moving male Sprague Dawley rats. We investigated how, across three brain regions, EEG features are correlated with EIB modulation. We found that the periodic component was composed of seven frequency bands that presented region-dependent and compound-dependent changes. The aperiodic component was also different between compounds and brain regions. Importantly, the parametrization into periodic and aperiodic components unveiled correlations between quantitative EEG and plasma concentrations of pharmacological compounds. MK-801 exposures were positively correlated with the slope of the aperiodic component. Concerning the periodic component, MK-801 exposures correlated negatively with the peak frequency of low-γ oscillations but positively with those of high-γ and high-frequency oscillations (HFOs). As for the power, θ and low-γ oscillations correlated negatively with MK-801, whereas mid-γ correlated positively. Diazepam correlated negatively with the knee of the aperiodic component, positively to β and negatively to low-γ oscillatory power, and positively to the modal frequency of θ, low-γ, mid-γ, and high-γ. In conclusion, correlations between exposures and pharmacodynamic effects can be better-understood thanks to the parametrization of EEG into periodic and aperiodic components. Such parametrization could be key in functional biomarker discovery.
Collapse
Affiliation(s)
- Irene Gonzalez-Burgos
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
- Program of Neuroscience, Centro de Investigación Médica Aplicada, Universidad de Navarra, Pamplona 31080, Spain
- Instituto de Investigación Sanitaria de Navarra (Navarra Institute for Health Research), Pamplona 31080, Spain
| | - Marie Bainier
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Simon Gross
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
| | - Philipp Schoenenberger
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
| | - José A Ochoa
- Program of Neuroscience, Centro de Investigación Médica Aplicada, Universidad de Navarra, Pamplona 31080, Spain
- Instituto de Investigación Sanitaria de Navarra (Navarra Institute for Health Research), Pamplona 31080, Spain
| | - Miguel Valencia
- Program of Neuroscience, Centro de Investigación Médica Aplicada, Universidad de Navarra, Pamplona 31080, Spain
- Instituto de Investigación Sanitaria de Navarra (Navarra Institute for Health Research), Pamplona 31080, Spain
- Institute of Data Science and Artificial Intelligence, Universidad de Navarra, Pamplona, Spain
| | - Roger L Redondo
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, Basel 4070, Switzerland
| |
Collapse
|
10
|
Gozzi A, Zerbi V. Modeling Brain Dysconnectivity in Rodents. Biol Psychiatry 2023; 93:419-429. [PMID: 36517282 DOI: 10.1016/j.biopsych.2022.09.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/19/2022] [Accepted: 09/10/2022] [Indexed: 02/04/2023]
Abstract
Altered or atypical functional connectivity as measured with functional magnetic resonance imaging (fMRI) is a hallmark feature of brain connectopathy in psychiatric, developmental, and neurological disorders. However, the biological underpinnings and etiopathological significance of this phenomenon remain unclear. The recent development of MRI-based techniques for mapping brain function in rodents provides a powerful platform to uncover the determinants of functional (dys)connectivity, whether they are genetic mutations, environmental risk factors, or specific cellular and circuit dysfunctions. Here, we summarize the recent contribution of rodent fMRI toward a deeper understanding of network dysconnectivity in developmental and psychiatric disorders. We highlight substantial correspondences in the spatiotemporal organization of rodent and human fMRI networks, supporting the translational relevance of this approach. We then show how this research platform might help us comprehend the importance of connectional heterogeneity in complex brain disorders and causally relate multiscale pathogenic contributors to functional dysconnectivity patterns. Finally, we explore how perturbational techniques can be used to dissect the fundamental aspects of fMRI coupling and reveal the causal contribution of neuromodulatory systems to macroscale network activity, as well as its altered dynamics in brain diseases. These examples outline how rodent functional imaging is poised to advance our understanding of the bases and determinants of human functional dysconnectivity.
Collapse
Affiliation(s)
- Alessandro Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto, Italy.
| | - Valerio Zerbi
- Neuro-X Institute, School of Engineering, École polytechnique fédérale de Lausanne, Lausanne, Switzerland; CIBM Center for Biomedical Imaging, Lausanne, Switzerland.
| |
Collapse
|
11
|
The effect of ketamine and D-cycloserine on the high frequency resting EEG spectrum in humans. Psychopharmacology (Berl) 2023; 240:59-75. [PMID: 36401646 PMCID: PMC9816261 DOI: 10.1007/s00213-022-06272-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 10/28/2022] [Indexed: 11/21/2022]
Abstract
RATIONALE Preclinical studies indicate that high-frequency oscillations, above 100 Hz (HFO:100-170 Hz), are a potential translatable biomarker for pharmacological studies, with the rapid acting antidepressant ketamine increasing both gamma (40-100 Hz) and HFO. OBJECTIVES To assess the effect of the uncompetitive NMDA antagonist ketamine, and of D-cycloserine (DCS), which acts at the glycine site on NMDA receptors on HFO in humans. METHODS We carried out a partially double-blind, 4-way crossover study in 24 healthy male volunteers. Each participant received an oral tablet and an intravenous infusion on each of four study days. The oral treatment was either DCS (250 mg or 1000 mg) or placebo. The infusion contained 0.5 mg/kg ketamine or saline placebo. The four study conditions were therefore placebo-placebo, 250 mg DCS-placebo, 1000 mg DCS-placebo, or placebo-ketamine. RESULTS Compared with placebo, frontal midline HFO magnitude was increased by ketamine (p = 0.00014) and 1000 mg DCS (p = 0.013). Frontal gamma magnitude was also increased by both these treatments. However, at a midline parietal location, only HFO were increased by DCS, and not gamma, whilst ketamine increased both gamma and HFO at this location. Ketamine induced psychomimetic effects, as measured by the PSI scale, whereas DCS did not increase the total PSI score. The perceptual distortion subscale scores correlated with the posterior low gamma to frontal high beta ratio. CONCLUSIONS Our results suggest that, at high doses, a partial NMDA agonist (DCS) has similar effects on fast neural oscillations as an NMDA antagonist (ketamine). As HFO were induced without psychomimetic effects, they may prove a useful drug development target.
Collapse
|
12
|
English BA, Ereshefsky L. Experimental Medicine Approaches in Early-Phase CNS Drug Development. ADVANCES IN NEUROBIOLOGY 2023; 30:417-455. [PMID: 36928860 DOI: 10.1007/978-3-031-21054-9_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
Traditionally, Phase 1 clinical trials were largely conducted in healthy normal volunteers and focused on collection of safety, tolerability, and pharmacokinetic data. However, in the CNS therapeutic area, with more drugs failing in later phase development, Phase 1 trials have undergone an evolution that includes incorporation of novel approaches involving novel study designs, inclusion of biomarkers, and early inclusion of patients to improve the pharmacologic understanding of novel CNS-active compounds early in clinical development with the hope of improving success in later phase pivotal trials. In this chapter, the authors will discuss the changing landscape of Phase 1 clinical trials in CNS, including novel trial methodology, inclusion of pharmacodynamic biomarkers, and experimental medicine approaches to inform early decision-making in clinical development.
Collapse
|
13
|
Teal LB, Ingram SM, Bubser M, McClure E, Jones CK. The Evolving Role of Animal Models in the Discovery and Development of Novel Treatments for Psychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2023; 30:37-99. [PMID: 36928846 DOI: 10.1007/978-3-031-21054-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Historically, animal models have been routinely used in the characterization of novel chemical entities (NCEs) for various psychiatric disorders. Animal models have been essential in the in vivo validation of novel drug targets, establishment of lead compound pharmacokinetic to pharmacodynamic relationships, optimization of lead compounds through preclinical candidate selection, and development of translational measures of target occupancy and functional target engagement. Yet, with decades of multiple NCE failures in Phase II and III efficacy trials for different psychiatric disorders, the utility and value of animal models in the drug discovery process have come under intense scrutiny along with the widespread withdrawal of the pharmaceutical industry from psychiatric drug discovery. More recently, the development and utilization of animal models for the discovery of psychiatric NCEs has undergone a dynamic evolution with the application of the Research Domain Criteria (RDoC) framework for better design of preclinical to clinical translational studies combined with innovative genetic, neural circuitry-based, and automated testing technologies. In this chapter, the authors will discuss this evolving role of animal models for improving the different stages of the discovery and development in the identification of next generation treatments for psychiatric disorders.
Collapse
Affiliation(s)
- Laura B Teal
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Shalonda M Ingram
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Michael Bubser
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Elliott McClure
- College of Pharmacy and Health Sciences, Lipscomb University, Nashville, TN, USA
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
14
|
Carmichael O. The Role of fMRI in Drug Development: An Update. ADVANCES IN NEUROBIOLOGY 2023; 30:299-333. [PMID: 36928856 DOI: 10.1007/978-3-031-21054-9_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Functional magnetic resonance imaging (fMRI) of the brain is a technology that holds great potential for increasing the efficiency of drug development for the central nervous system (CNS). In preclinical studies and both early- and late-phase human trials, fMRI has the potential to improve cross-species translation of drug effects, help to de-risk compounds early in development, and contribute to the portfolio of evidence for a compound's efficacy and mechanism of action. However, to date, the utilization of fMRI in the CNS drug development process has been limited. The purpose of this chapter is to explore this mismatch between potential and utilization. This chapter provides introductory material related to fMRI and drug development, describes what is required of fMRI measurements for them to be useful in a drug development setting, lists current capabilities of fMRI in this setting and challenges faced in its utilization, and ends with directions for future development of capabilities in this arena. This chapter is the 5-year update of material from a previously published workshop summary (Carmichael et al., Drug DiscovToday 23(2):333-348, 2018).
Collapse
Affiliation(s)
- Owen Carmichael
- Pennington Biomedical Research Center, Baton Rouge, LA, USA.
| |
Collapse
|
15
|
Kawazoe K, McGlynn R, Felix W, Sevilla R, Liao S, Kulkarni P, Ferris CF. Dose-dependent effects of esketamine on brain activity in awake mice: A BOLD phMRI study. Pharmacol Res Perspect 2022; 10:e01035. [PMID: 36504448 PMCID: PMC9743060 DOI: 10.1002/prp2.1035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 12/14/2022] Open
Abstract
Pharmacological magnetic resonance imaging (phMRI) is a noninvasive method used to evaluate neural circuitry involved in the behavioral effects of drugs like ketamine, independent of their specific biochemical mechanism. The study was designed to evaluate the immediate effect of esketamine, the S-isomer of (±) ketamine on brain activity in awake mice using blood oxygenation level dependent (BOLD) imaging. It was hypothesized the prefrontal cortex, hippocampus, and brain areas associated with reward and motivation would show a dose-dependent increase in brain activity. Mice were given vehicle, 1.0, 3.3, or 10 mg/kg esketamine I.P. and imaged for 10 min post-treatment. Data for each treatment were registered to a 3D MRI mouse brain atlas providing site-specific information on 134 different brain areas. There was a global change in brain activity for both positive and negative BOLD signal affecting over 50 brain areas. Many areas showed a dose-dependent decrease in positive BOLD signal, for example, cortex, hippocampus, and thalamus. The most common profile when comparing the three doses was a U-shape with the 3.3 dose having the lowest change in signal. At 1.0 mg/kg there was a significant increase in positive BOLD in forebrain areas and hippocampus. The anticipated dose-dependent increase in BOLD was not realized; instead, the lowest dose of 1.0 mg/kg had the greatest effect on brain activity. The prefrontal cortex and hippocampus were significantly activated corroborating previous imaging studies in humans and animals. The unexpected sensitivity to the 1.0 mg/kg dose of esketamine could be explained by imaging in fully awake mice without the confound of anesthesia and/or its greater affinity for the N-methyl-d-aspartate receptor (NMDAR) receptor than (±) ketamine.
Collapse
Affiliation(s)
- Kyrsten Kawazoe
- Department of Pharmaceutical SciencesNortheastern UniversityBostonMassachusettsUSA
| | - Ryan McGlynn
- Department of Pharmaceutical SciencesNortheastern UniversityBostonMassachusettsUSA
| | - Wilder Felix
- Department of Pharmaceutical SciencesNortheastern UniversityBostonMassachusettsUSA
| | - Raquel Sevilla
- Department of Pharmaceutical SciencesNortheastern UniversityBostonMassachusettsUSA
| | - Siyang Liao
- Department of Pharmaceutical SciencesNortheastern UniversityBostonMassachusettsUSA
| | - Praveen Kulkarni
- Center for Translational NeuroimagingNortheastern UniversityMassachusettsBostonUSA
| | - Craig F. Ferris
- Department of Pharmaceutical SciencesNortheastern UniversityBostonMassachusettsUSA
- Center for Translational NeuroimagingNortheastern UniversityMassachusettsBostonUSA
- Department of PsychologyNortheastern UniversityBostonMassachusettsUSA
| |
Collapse
|
16
|
Gärtner M, de Rover M, Václavů L, Scheidegger M, van Osch MJP, Grimm S. Increase in thalamic cerebral blood flow is associated with antidepressant effects of ketamine in major depressive disorder. World J Biol Psychiatry 2022; 23:643-652. [PMID: 34985394 DOI: 10.1080/15622975.2021.2020900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Ketamine is a promising treatment option for patients with Major Depressive Disorder (MDD) and has become an important research tool to investigate antidepressant mechanisms of action. However, imaging studies attempting to characterise ketamine's mechanism of action using blood oxygen level-dependent signal (BOLD) imaging have yielded inconsistent results- at least partly due to intrinsic properties of the BOLD contrast, which measures a complex signal related to neural activity. To circumvent the limitations associated with the BOLD signal, we used arterial spin labelling (ASL) as an unambiguous marker of neuronal activity-related changes in cerebral blood flow (CBF). We measured CBF in 21 MDD patients at baseline and 24 h after receiving a single intravenous infusion of subanesthetic ketamine and examined relationships with clinical outcomes. Our findings demonstrate that increase in thalamus perfusion 24 h after ketamine administration is associated with greater improvement of depressive symptoms. Furthermore, lower thalamus perfusion at baseline is associated both with larger increases in perfusion 24 h after ketamine administration and with stronger reduction of depressive symptoms. These findings indicate that ASL is not only a useful tool to broaden our understanding of ketamine's mechanism of action but might also have the potential to inform treatment decisions based on CBF-defined regional disruptions.
Collapse
Affiliation(s)
- Matti Gärtner
- MSB-Medical School Berlin, Berlin, Germany.,Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Mischa de Rover
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands.,Department of Clinical Psychology, Institute of Psychology, Leiden University, Leiden, Netherlands
| | - Lena Václavů
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, Netherlands
| | - Milan Scheidegger
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| | - Matthias J P van Osch
- Department of Radiology, C.J. Gorter Center for High Field MRI, Leiden University Medical Center, Leiden, Netherlands
| | - Simone Grimm
- MSB-Medical School Berlin, Berlin, Germany.,Department of Psychiatry and Psychotherapy, Campus Benjamin Franklin, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Zurich, Switzerland
| |
Collapse
|
17
|
The Effects of Acute Δ 9-Tetrahydrocannabinol on Striatal Glutamatergic Function: A Proton Magnetic Resonance Spectroscopy Study. BIOLOGICAL PSYCHIATRY: COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2021; 6:660-667. [PMID: 34099186 DOI: 10.1016/j.bpsc.2021.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Cannabis and its main psychoactive component, Δ9-tetrahydrocannabinol (THC), can elicit transient psychotic symptoms. A key candidate biological mechanism of how THC induces psychotic symptoms is the modulation of glutamate in the brain. We sought to investigate the effects of acute THC administration on striatal glutamate levels and its relationship to the induction of psychotic symptoms. METHODS We used proton magnetic resonance spectroscopy to measure glutamate levels in the striatum in 20 healthy participants after THC (15 mg, oral) and matched placebo administration in a randomized, double-blind, placebo-controlled design. Psychotic symptoms were measured using the Psychotomimetic States Inventory. RESULTS We found that THC administration did not significantly change glutamate (glutamate plus glutamine relative to creatine) concentration in the striatum (p = .58; scaled Jeffreys-Zellner-Siow Bayes factor = 4.29). THC increased psychotic symptoms, but the severity of these symptoms was not correlated with striatal glutamate levels. CONCLUSIONS These findings suggest that oral administration of 15 mg of THC does not result in altered striatal glutamate levels. Further work is needed to clarify the effects of THC on striatal glutamate.
Collapse
|
18
|
Grabb MC, Hillefors M, Potter WZ. The NIMH 'Fast-Fail Trials' (FAST) Initiative: Rationale, Promise, and Progress. Pharmaceut Med 2021; 34:233-245. [PMID: 32705569 DOI: 10.1007/s40290-020-00343-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In 2012, the US National Institute of Mental Health launched three clinical trial contracts under a new FAST initiative. The overall goal for these contracts (Fast-Fail Trials) was to focus early-stage trials, testing novel pharmacologic agents that target the central nervous system, on pharmacologic-based designs to objectively identify doses that produce central nervous system effects. The three contracts targeted different psychiatric populations: psychotic (FAST-PS), mood and anxiety (FAST-MAS), and autism spectrum disorders (FAST-AS). The FAST initiative was a first attempt for the National Institute of Mental Health to adapt an experimental medicine approach to its clinical trial portfolio. As the Fast-Fail trials implemented this new approach for the field, we present the rationale for each trial, design considerations, results, and how each one contributed new knowledge to the field of psychopharmacology; important lessons for pharma and biotech. Under the FAST initiative, the National Institute of Mental Health assembled research teams with a broad range of expertise, who developed and validated the outcome measures and study protocol, and conducted multi-site clinical trials, testing candidate compounds. In the FAST-PS contract, the team validated an imaging-based pharmacodynamic biomarker of the effect of ketamine in the brain that could be utilized in subsequent clinical trials. The initial FAST-AS study was an important first step in the design of early-stage target-engagement trials in autism spectrum disorder, suggesting that a resting electroencephalogram can be used as a pharmacodynamic measure in future studies. The FAST-MAS study showed that blocking the kappa-opioid receptor significantly affects functional magnetic resonance imaging ventral striatal activation in the monetary incentive delay task in anticipation of gain. Together, the outcomes of the FAST-FAIL trials demonstrated the importance of rigorously designed and informative central nervous system trials, including the value of pharmacodynamic measures in early-stage trials. Use of these measures furthered our knowledge about the relationship between specific molecular mechanisms, brain effects, and therapeutic effects in patients with mental illnesses.
Collapse
Affiliation(s)
- Margaret C Grabb
- National Institutes of Health, National Institute of Mental Health, 6001 Executive Boulevard, Bethesda, MD, 20892, USA.
| | - Mi Hillefors
- National Institutes of Health, National Institute of Mental Health, 6001 Executive Boulevard, Bethesda, MD, 20892, USA
| | - William Z Potter
- National Institutes of Health, National Institute of Mental Health, 6001 Executive Boulevard, Bethesda, MD, 20892, USA
| |
Collapse
|
19
|
Imaging of the dopamine system with focus on pharmacological MRI and neuromelanin imaging. Eur J Radiol 2021; 140:109752. [PMID: 34004428 DOI: 10.1016/j.ejrad.2021.109752] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/26/2021] [Accepted: 04/29/2021] [Indexed: 11/21/2022]
Abstract
The dopamine system in the brain is involved in a variety of neurologic and psychiatric disorders, such as Parkinson's disease, attention-deficit/hyperactivity disorder and psychosis. Different aspects of the dopamine system can be visualized and measured with positron emission tomography (PET) and single photon emission computed tomography (SPECT), including dopamine receptors, dopamine transporters, and dopamine release. New developments in MR imaging also provide proxy measures of the dopamine system in the brain, offering alternatives with the advantages MR imaging, i.e. no radiation, lower costs, usually less invasive and time consuming. This review will give an overview of these developments with a focus on the most developed techniques: pharmacological MRI (phMRI) and neuromelanin sensitive MRI (NM-MRI). PhMRI is a collective term for functional MRI techniques that administer a pharmacological challenge to assess its effects on brain hemodynamics. By doing so, it indirectly assesses brain neurotransmitter function such as dopamine function. NM-MRI is an upcoming MRI technique that enables in vivo visualization and semi-quantification of neuromelanin in the substantia nigra. Neuromelanin is located in the cell bodies of dopaminergic neurons of the nigrostriatal pathway and can be used as a proxy measure for long term dopamine function or degeneration of dopaminergic neurons. Both techniques are still primarily used in clinical research, but there is promise for clinical application, in particular for NM-MRI in dopaminergic neurodegenerative diseases like Parkinson's disease.
Collapse
|
20
|
Alexander L, Jelen LA, Mehta MA, Young AH. The anterior cingulate cortex as a key locus of ketamine's antidepressant action. Neurosci Biobehav Rev 2021; 127:531-554. [PMID: 33984391 DOI: 10.1016/j.neubiorev.2021.05.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 12/30/2022]
Abstract
The subdivisions of the anterior cingulate cortex (ACC) - including subgenual, perigenual and dorsal zones - are implicated in the etiology, pathogenesis and treatment of major depression. We review an emerging body of evidence which suggests that changes in ACC activity are critically important in mediating the antidepressant effects of ketamine, the prototypical member of an emerging class of rapidly acting antidepressants. Infusions of ketamine induce acute (over minutes) and post-acute (over hours to days) modulations in subgenual and perigenual activity, and importantly, these changes can correlate with antidepressant efficacy. The subgenual and dorsal zones of the ACC have been specifically implicated in ketamine's anti-anhedonic effects. We emphasize the synergistic relationship between neuroimaging studies in humans and brain manipulations in animals to understand the causal relationship between changes in brain activity and therapeutic efficacy. We conclude with circuit-based perspectives on ketamine's action: first, related to ACC function in a central network mediating affective pain, and second, related to its role as the anterior node of the default mode network.
Collapse
Affiliation(s)
- Laith Alexander
- Department of Psychological Medicine, School of Academic Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; St Thomas' Hospital, London, United Kingdom.
| | - Luke A Jelen
- Department of Psychological Medicine, School of Academic Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - Mitul A Mehta
- Department of Psychological Medicine, School of Academic Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Allan H Young
- Department of Psychological Medicine, School of Academic Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; South London and Maudsley NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
21
|
The M1/M4 preferring muscarinic agonist xanomeline modulates functional connectivity and NMDAR antagonist-induced changes in the mouse brain. Neuropsychopharmacology 2021; 46:1194-1206. [PMID: 33342996 PMCID: PMC8115158 DOI: 10.1038/s41386-020-00916-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/02/2020] [Accepted: 11/10/2020] [Indexed: 12/25/2022]
Abstract
Cholinergic drugs acting at M1/M4 muscarinic receptors hold promise for the treatment of symptoms associated with brain disorders characterized by cognitive impairment, mood disturbances, or psychosis, such as Alzheimer's disease or schizophrenia. However, the brain-wide functional substrates engaged by muscarinic agonists remain poorly understood. Here we used a combination of pharmacological fMRI (phMRI), resting-state fMRI (rsfMRI), and resting-state quantitative EEG (qEEG) to investigate the effects of a behaviorally active dose of the M1/M4-preferring muscarinic agonist xanomeline on brain functional activity in the rodent brain. We investigated both the effects of xanomeline per se and its modulatory effects on signals elicited by the NMDA-receptor antagonists phencyclidine (PCP) and ketamine. We found that xanomeline induces robust and widespread BOLD signal phMRI amplitude increases and decreased high-frequency qEEG spectral activity. rsfMRI mapping in the mouse revealed that xanomeline robustly decreased neocortical and striatal connectivity but induces focal increases in functional connectivity within the nucleus accumbens and basal forebrain. Notably, xanomeline pre-administration robustly attenuated both the cortico-limbic phMRI response and the fronto-hippocampal hyper-connectivity induced by PCP, enhanced PCP-modulated functional connectivity locally within the nucleus accumbens and basal forebrain, and reversed the gamma and high-frequency qEEG power increases induced by ketamine. Collectively, these results show that xanomeline robustly induces both cholinergic-like neocortical activation and desynchronization of functional networks in the mammalian brain. These effects could serve as a translatable biomarker for future clinical investigations of muscarinic agents, and bear mechanistic relevance for the putative therapeutic effect of these class of compounds in brain disorders.
Collapse
|
22
|
Woelfer M, Li M, Colic L, Liebe T, Di X, Biswal B, Murrough J, Lessmann V, Brigadski T, Walter M. Ketamine-induced changes in plasma brain-derived neurotrophic factor (BDNF) levels are associated with the resting-state functional connectivity of the prefrontal cortex. World J Biol Psychiatry 2020; 21:696-710. [PMID: 31680600 DOI: 10.1080/15622975.2019.1679391] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Synaptic plasticity and brain-derived neurotrophic factor (BDNF) signalling are proposed to play key roles in antidepressant drug action. Ketamine, an N-methyl-D-aspartate receptor antagonist and putative antidepressant, may increase synaptic plasticity in prefrontal cortex through higher expression of BDNF. Furthermore, ketamine was shown to change resting-state functional connectivity (RSFC) of dorsomedial prefrontal cortex (dmPFC). METHODS In a randomised, placebo-controlled study, we investigated acutely (100 min) and at 24 h following subanesthetic ketamine infusion which dmPFC seeded RSFC changes are most strongly associated with plasma BDNF level changes in 53 healthy participants (21 females, age: 24.4 ± 2.9 years) using 7 T-fMRI. RESULTS We observed higher relative levels of BDNF 2 h and 24 h after ketamine compared to placebo. Whole-brain regression revealed that the change in BDNF after 24 h was associated with RSFC decreases from dmPFC to posterior cingulate cortex and ventromedial PFC at 24 h and exploratively also at the 100 min measurement point. Follow-up analyses revealed that RSFC reductions following ketamine were restricted to subjects showing increased BDNF levels at 24 h. CONCLUSIONS Our findings indicate BDNF level dynamics following ketamine are related to acute and 24 h RSFC changes. Particularly when BDNF increases are observed after ketamine infusion, a disconnection from dmPFC after 24 h is seen and may reflect synaptic plasticity effects.
Collapse
Affiliation(s)
- Marie Woelfer
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany.,Leibniz Institute for Neurobiology, Magdeburg, Germany.,Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Meng Li
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany.,Department of Psychiatry and Psychotherapy, University Tuebingen, Tuebingen, Germany
| | - Lejla Colic
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany.,Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Thomas Liebe
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany
| | - Xin Di
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Bharat Biswal
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA.,School of Life Sciences and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - James Murrough
- Depression and Anxiety Center for Discovery and Treatment, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Volkmar Lessmann
- Institute of Physiology, Otto-von-Guericke-University, Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Tanja Brigadski
- Institute of Physiology, Otto-von-Guericke-University, Magdeburg, Germany.,Department of Informatics and Microsystems Technology, University of Applied Science Kaiserslautern, Zweibrücken, Germany
| | - Martin Walter
- Clinical Affective Neuroimaging Laboratory (CANLAB), Magdeburg, Germany.,Department of Psychiatry and Psychotherapy, University Tuebingen, Tuebingen, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| |
Collapse
|
23
|
Kantrowitz JT, Grinband J, Goff DC, Lahti AC, Marder SR, Kegeles LS, Girgis RR, Sobeih T, Wall MM, Choo TH, Green MF, Yang YS, Lee J, Horga G, Krystal JH, Potter WZ, Javitt DC, Lieberman JA. Proof of mechanism and target engagement of glutamatergic drugs for the treatment of schizophrenia: RCTs of pomaglumetad and TS-134 on ketamine-induced psychotic symptoms and pharmacoBOLD in healthy volunteers. Neuropsychopharmacology 2020; 45:1842-1850. [PMID: 32403118 PMCID: PMC7608251 DOI: 10.1038/s41386-020-0706-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/12/2020] [Accepted: 05/06/2020] [Indexed: 12/15/2022]
Abstract
Glutamate neurotransmission is a prioritized target for antipsychotic drug development. Two metabotropic glutamate receptor 2/3 (mGluR2/3) agonists (pomaglumetad [POMA] and TS-134) were assessed in two Phase Ib proof of mechanism studies of comparable designs and using identical clinical assessments and pharmacoBOLD methodology. POMA was examined in a randomized controlled trial under double-blind conditions for 10-days at doses of 80 or 320 mg/d POMA versus placebo (1:1:1 ratio). The TS-134 trial was a randomized, single-blind, 6-day study of 20 or 60 mg/d TS-134 versus placebo (5:5:2 ratio). Primary outcomes were ketamine-induced changes in pharmacoBOLD in the dorsal anterior cingulate cortex (dACC) and symptoms reflected on the Brief Psychiatric Rating Scale (BPRS). Both trials were conducted contemporaneously. 95 healthy volunteers were randomized to POMA and 63 to TS-134. High-dose POMA significantly reduced ketamine-induced BPRS total symptoms within and between-groups (p < 0.01, d = -0.41; p = 0.04, d = -0.44, respectively), but neither POMA dose significantly suppressed ketamine-induced dACC pharmacoBOLD. In contrast, low-dose TS-134 led to moderate to large within and between group reductions in both BPRS positive symptoms (p = 0.02, d = -0.36; p = 0.008, d = -0.82, respectively) and dACC pharmacoBOLD (p = 0.004, d = -0.56; p = 0.079, d = -0.50, respectively) using pooled across-study placebo data. High-dose POMA exerted significant effects on clinical symptoms, but not on target engagement, suggesting a higher dose may yet be needed, while the low dose of TS-134 showed evidence of symptom reduction and target engagement. These results support further investigation of mGluR2/3 and other glutamate-targeted treatments for schizophrenia.
Collapse
Affiliation(s)
- Joshua T. Kantrowitz
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA ,grid.250263.00000 0001 2189 4777Nathan Kline Institute, Orangeburg, NY USA
| | - Jack Grinband
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| | - Donald C. Goff
- grid.250263.00000 0001 2189 4777Nathan Kline Institute, Orangeburg, NY USA ,grid.240324.30000 0001 2109 4251NYU Langone Medical Center, New York, NY USA
| | - Adrienne C. Lahti
- grid.265892.20000000106344187University of Alabama at Birmingham, Birmingham, AL USA
| | | | - Lawrence S. Kegeles
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| | - Ragy R. Girgis
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| | - Tarek Sobeih
- grid.250263.00000 0001 2189 4777Nathan Kline Institute, Orangeburg, NY USA
| | - Melanie M. Wall
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| | - Tse-Hwei Choo
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| | | | - Yvonne S. Yang
- grid.19006.3e0000 0000 9632 6718UCLA, Los Angeles, CA USA
| | - Junghee Lee
- grid.19006.3e0000 0000 9632 6718UCLA, Los Angeles, CA USA
| | - Guillermo Horga
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| | - John H. Krystal
- grid.47100.320000000419368710Yale University School of Medicine, New Haven, CT USA
| | - William Z. Potter
- grid.94365.3d0000 0001 2297 5165National Institutes of Health, Bethesda, MD USA
| | - Daniel C. Javitt
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA ,grid.250263.00000 0001 2189 4777Nathan Kline Institute, Orangeburg, NY USA
| | - Jeffrey A. Lieberman
- grid.21729.3f0000000419368729Columbia University, New York, NY USA ,grid.413734.60000 0000 8499 1112New York State Psychiatric Institute, New York, NY USA
| |
Collapse
|
24
|
McMillan R, Muthukumaraswamy SD. The neurophysiology of ketamine: an integrative review. Rev Neurosci 2020; 31:457-503. [DOI: 10.1515/revneuro-2019-0090] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 01/26/2020] [Indexed: 12/13/2022]
Abstract
AbstractThe drug ketamine has been extensively studied due to its use in anaesthesia, as a model of psychosis and, most recently, its antidepressant properties. Understanding the physiology of ketamine is complex due to its rich pharmacology with multiple potential sites at clinically relevant doses. In this review of the neurophysiology of ketamine, we focus on the acute effects of ketamine in the resting brain. We ascend through spatial scales starting with a complete review of the pharmacology of ketamine and then cover its effects on in vitro and in vivo electrophysiology. We then summarise and critically evaluate studies using EEG/MEG and neuroimaging measures (MRI and PET), integrating across scales where possible. While a complicated and, at times, confusing picture of ketamine’s effects are revealed, we stress that much of this might be caused by use of different species, doses, and analytical methodologies and suggest strategies that future work could use to answer these problems.
Collapse
Affiliation(s)
- Rebecca McMillan
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Suresh D. Muthukumaraswamy
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| |
Collapse
|
25
|
Intragastric fructose administration interacts with emotional state in homeostatic and hedonic brain regions. Nutr Neurosci 2020; 25:581-592. [PMID: 32558624 DOI: 10.1080/1028415x.2020.1781326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Background: Interoceptive properties of food may influence emotional state and its neural basis, as shown for fatty acids but remains unstudied for carbohydrates.Objectives: To study the effects of fructose and its interaction with sad emotion on brain activity in homeostatic and hedonic regions and investigate whether gut hormone responses can explain effects.Design: In 15 healthy subjects, brain activity for 40min after intragastric infusion of fructose (25g) or water was recorded using a cross-over pharmacological magnetic resonance imaging (phMRI) paradigm. Sad or neutral emotional states were induced by classical music and emotional facial expressions. Emotional state was assessed using the Self-Assessment Manikin. Blood samples were taken to assess gut hormone levels. Brain responses to fructose versus placebo, sad versus neutral emotion, and their interaction were analyzed over time in a single mask of a priori defined regions of interest at a voxel-level threshold of pFWEcorrected <0.05. Effects on emotion and hormones were tested using linear mixed models.Results: No main effects of fructose, emotion, or fructose-by-emotion interaction on emotional ratings were observed. Main effects of fructose, emotion and aninteraction effect were found on brain activity (medulla, midbrain, hypothalamus, basal ganglia, anterior insula, orbitofrontal cortex, anterior cingulate cortex and amygdala). An increase in circulating GLP-1 after fructose in neutral emotion was abolished during sad emotion (fructose-by-emotion-by-time, p=0.041). Ghrelin levels were higher in sad emotion (time-by-emotion, p=0.037).Conclusions: Emotional state interacts with brain and endocrine responses to intragastric infusion of 25 g of fructose, however such an effect was not found at behavioral level.Trial registration: ClinicalTrials.gov identifier: NCT02946983.
Collapse
|
26
|
Examining fMRI time-series entropy as a marker for brain E/I balance with pharmacological neuromodulation in a non-human primate translational model. Neurosci Lett 2020; 728:134984. [PMID: 32315710 DOI: 10.1016/j.neulet.2020.134984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 04/02/2020] [Accepted: 04/13/2020] [Indexed: 11/23/2022]
Abstract
Recently, there has been a lot of interest in the neuroimaging community in exploring fMRI time-series measures of local neuronal activity and excitation/inhibition (E/I) balance in the brain. In this preliminary study we probed the sensitivity of widely used sample entropy (SE) measure at multiple scales to controlled alteration of the brain's E/I balance in non-human primates (NHPs) with a well-characterized sub-anesthetic ketamine infusion fMRI model. We found that SE failed to detect the expected changes in E/I balance induced by ketamine. Subsequently, noticing that the complexity in the time series contributing SE could be dominated by non-neuronal noise in this experimental setting, we developed a new time-series measure called restricted sample entropy (RSE) by restricting SE estimations to regular portions of the fMRI time-series. RSE was able to adequately reflect the increased excitatory activity engendered by disinhibition of glutamergic neurons, through sub-anesthetic ketamine infusion. These results show that RSE is potentially a powerful tool for examining local neural activity, E/I balance, and alterations in brain state.
Collapse
|
27
|
Hindocha C, Quattrone D, Freeman TP, Murray RM, Mondelli V, Breen G, Curtis C, Morgan CJA, Valerie Curran H, Di Forti M. Do AKT1, COMT and FAAH influence reports of acute cannabis intoxication experiences in patients with first episode psychosis, controls and young adult cannabis users? Transl Psychiatry 2020; 10:143. [PMID: 32398646 PMCID: PMC7217850 DOI: 10.1038/s41398-020-0823-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 04/07/2020] [Accepted: 04/28/2020] [Indexed: 12/22/2022] Open
Abstract
Epidemiological and biological evidence support the association between heavy cannabis use and psychosis. However, it is unclear which cannabis users are susceptible to its psychotogenic effect. Therefore, understanding genetic factors contributing to this relationship might prove an important strategy to identify the mechanisms underlying cannabis-associated psychotic experiences. We aimed to determine how variation in AKT1, COMT and FAAH genotypes, and their interaction with three different groups (first episode psychosis (FEP) patients (n = 143), controls (n = 92) and young adult (YA) cannabis users n = 485)) influenced cannabis experiences, in those who had used cannabis at least once. We investigated the role of AKT1 (rs2494732), COMT Val158Met (rs4680) and FAAH (rs324420) on cannabis experiences by combining data from a large case-control study of FEP patients, with a naturalistic study of YA cannabis users (n = 720). Outcome measures were cannabis-induced psychotic-like experiences (cPLEs) and euphoric experiences (cEEs). We used linear mixed effects models to assess the effects of each genotype and their interaction with group, adjusting for age, sex, ethnicity, age of first cannabis use, years of use and frequency. cPLEs were more frequent in FEP patients than controls and YA cannabis users. cEEs were more prevalent in YA cannabis users than FEP patients or controls. Variation in AKT1, COMT or FAAH was not associated with cPLEs/cEEs. There was no interaction between genotype and group (FEP cases, controls and YA cannabis users) on cPLEs/cEEs. In conclusion, AKT1, COMT or FAAH did not modulate specific psychotomimetic response to cannabis and did not interact with group, contrary to previous research.
Collapse
Affiliation(s)
- Chandni Hindocha
- Clinical Psychopharmacology Unit, Research Department of Clinical, Educational and Health Psychology, University College London, London, United Kingdom. .,Translational Psychiatry Research Group, Research Department of Mental Health Neuroscience, Division of Psychiatry, Faculty of Brain Sciences, University College London, London, United Kingdom. .,NIHR University College London Hospitals Biomedical Research Centre, University College Hospital, London, United Kingdom.
| | - Diego Quattrone
- grid.13097.3c0000 0001 2322 6764Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, SE5 8AF UK ,grid.451056.30000 0001 2116 3923National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King’s College London, London, UK ,grid.451052.70000 0004 0581 2008South London and Maudsley NHS Mental Health Foundation Trust, London, UK
| | - Tom P. Freeman
- grid.83440.3b0000000121901201Clinical Psychopharmacology Unit, Research Department of Clinical, Educational and Health Psychology, University College London, London, United Kingdom ,grid.83440.3b0000000121901201Translational Psychiatry Research Group, Research Department of Mental Health Neuroscience, Division of Psychiatry, Faculty of Brain Sciences, University College London, London, United Kingdom ,grid.7340.00000 0001 2162 1699Addiction and Mental Health Group (AIM), Department of Psychology, University of Bath, Bath, UK
| | - Robin M. Murray
- grid.451056.30000 0001 2116 3923National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King’s College London, London, UK ,grid.37640.360000 0000 9439 0839NIHR BioResource Centre Maudsley, NIHR Maudsley Biomedical Research Centre (BRC) at South London and Maudsley NHS Foundation Trust (SLaM), London, UK
| | - Valeria Mondelli
- grid.13097.3c0000 0001 2322 6764Department of Psychological Medicine, Institute of Psychiatry, Kings College London, De Crespigny Park, SE5 8AF London, UK
| | - Gerome Breen
- grid.13097.3c0000 0001 2322 6764Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, SE5 8AF UK ,grid.451056.30000 0001 2116 3923National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King’s College London, London, UK ,grid.13097.3c0000 0001 2322 6764Department of Psychosis Studies, Institute of Psychiatry, King’s College London, De Crespigny Park, Denmark Hill, London, SE5 8AF UK
| | - Charles Curtis
- grid.13097.3c0000 0001 2322 6764Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, SE5 8AF UK ,grid.451056.30000 0001 2116 3923National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King’s College London, London, UK ,grid.13097.3c0000 0001 2322 6764Department of Psychosis Studies, Institute of Psychiatry, King’s College London, De Crespigny Park, Denmark Hill, London, SE5 8AF UK
| | - Celia J. A. Morgan
- grid.83440.3b0000000121901201Clinical Psychopharmacology Unit, Research Department of Clinical, Educational and Health Psychology, University College London, London, United Kingdom ,grid.8391.30000 0004 1936 8024Psychopharmacology and Addiction Research Centre (PARC), University of Exeter, Exeter, UK
| | - H. Valerie Curran
- grid.83440.3b0000000121901201Clinical Psychopharmacology Unit, Research Department of Clinical, Educational and Health Psychology, University College London, London, United Kingdom ,grid.439749.40000 0004 0612 2754NIHR University College London Hospitals Biomedical Research Centre, University College Hospital, London, United Kingdom
| | - Marta Di Forti
- grid.13097.3c0000 0001 2322 6764Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, SE5 8AF UK ,grid.451056.30000 0001 2116 3923National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King’s College London, London, UK ,grid.451052.70000 0004 0581 2008South London and Maudsley NHS Mental Health Foundation Trust, London, UK
| |
Collapse
|
28
|
McMillan R, Sumner R, Forsyth A, Campbell D, Malpas G, Maxwell E, Deng C, Hay J, Ponton R, Sundram F, Muthukumaraswamy S. Simultaneous EEG/fMRI recorded during ketamine infusion in patients with major depressive disorder. Prog Neuropsychopharmacol Biol Psychiatry 2020; 99:109838. [PMID: 31843628 DOI: 10.1016/j.pnpbp.2019.109838] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 12/05/2019] [Accepted: 12/12/2019] [Indexed: 01/09/2023]
Abstract
A single subanaesthetic dose of ketamine rapidly alleviates the symptoms of major depressive disorder (MDD). However, few studies have investigated the acute effects of ketamine on the BOLD pharmacological magnetic resonance imaging (phMRI) response and EEG spectra. In a randomised, double-blind, active placebo-controlled crossover trial, resting-state simultaneous EEG/fMRI was collected during infusion of ketamine or active placebo (remifentanil) in 30 participants with MDD. Montgomery-Asberg depression rating scale scores showed a significant antidepressant effect of ketamine compared to placebo (69% response rate). phMRI analyses showed BOLD signal increases in the anterior cingulate and medial prefrontal cortices and sensitivity of the decrease in subgenual anterior cingulate cortex (sgACC) BOLD signal to noise correction. EEG spectral analysis showed increased theta, high beta, low and high gamma power, and decreased delta, alpha, and low beta power with differing time-courses. Low beta and high gamma power time courses explained significant variance in the BOLD signal. Interestingly, the variance explained by high gamma power was significantly associated with non-response to ketamine, but significant associations were not found for other neurophysiological markers when noise correction was implemented. The results suggest that the decrease in sgACC BOLD signal is potentially noise and unrelated to ketamine's antidepressant effect, highlighting the importance of noise correction and multiple temporal regressors for phMRI analyses. The lack of effects significantly associated with antidepressant response suggests the phMRI methodology employed was unable to detect such effects, the effect sizes are relatively small, or that other processes, e.g. neural plasticity, underlie ketamine's antidepressant effect.
Collapse
Affiliation(s)
| | | | - Anna Forsyth
- School of Pharmacy, University of Auckland, New Zealand
| | - Doug Campbell
- Department of Anaesthesiology, Auckland District Health Board, New Zealand
| | - Gemma Malpas
- Department of Anaesthesiology, Auckland District Health Board, New Zealand
| | - Elizabeth Maxwell
- Department of Anaesthesiology, Auckland District Health Board, New Zealand
| | - Carolyn Deng
- Department of Anaesthesiology, Auckland District Health Board, New Zealand
| | - John Hay
- Department of Anaesthesiology, Auckland District Health Board, New Zealand
| | - Rhys Ponton
- School of Pharmacy, University of Auckland, New Zealand
| | - Frederick Sundram
- Department of Psychological Medicine, University of Auckland, New Zealand
| | | |
Collapse
|
29
|
Higgins J, Barbieri E, Wang X, Mack J, Caplan D, Kiran S, Rapp B, Thompson C, Zinbarg R, Parrish T. Reliability of BOLD signals in chronic stroke-induced aphasia. Eur J Neurosci 2020; 52:3963-3978. [PMID: 32282965 DOI: 10.1111/ejn.14739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 11/30/2022]
Abstract
Investigating the neurobiology of language impairment and treatment in chronic stroke aphasia using fMRI requires an understanding of measurement variability within and between participants. In this multicenter study, we evaluated the scan-rescan reliability of an auditory and visual (written) story comprehension paradigm in stroke participants with aphasia (N = 65) and healthy controls (N = 22). The multi-modal task was conducted twice (~1 week apart) on separate visits upon study enrolment and twice again at completion three months later. A non-language visuomotor task was studied in the aphasia group only, which was conducted once per time point (3 months apart). While participants were asked to make responses during the comprehension task, these in-scanner responses were not recorded. Reliability was assessed using intraclass correlation coefficient (ICC) at both group and individual participant levels. The visual story comprehension condition had higher reliability than the auditory condition in both groups, with participants with aphasia exhibiting lower reliability than controls in both conditions (stroke ICC = .43, healthy ICC = .81). Differences in reliability within the group of participants with aphasia were found to be partially explained by overall language impairment as well as greater head motion. In the participants with aphasia, the visuomotor paradigm was found to have greater reliability than the story comprehension task at equivalent interscan intervals (visuomotor = 0.50, comprehension = 0.34), and its reliability was not associated with language impairment. This work highlights the importance of considering the reliability of fMRI tasks in aphasia research, provides strategies to improve reliability and has potential implications for the field of clinical neuroimaging in general.
Collapse
Affiliation(s)
- James Higgins
- Center for the Neurobiology of Language Recovery, Northwestern University, Evanston, IL, USA.,Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Elena Barbieri
- Center for the Neurobiology of Language Recovery, Northwestern University, Evanston, IL, USA.,Department of Communication Sciences and Disorders, School of Communication, Northwestern University, Evanston, IL, USA
| | - Xue Wang
- Center for the Neurobiology of Language Recovery, Northwestern University, Evanston, IL, USA.,Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jennifer Mack
- Center for the Neurobiology of Language Recovery, Northwestern University, Evanston, IL, USA.,Department of Communication Sciences and Disorders, School of Communication, Northwestern University, Evanston, IL, USA
| | - David Caplan
- Center for the Neurobiology of Language Recovery, Northwestern University, Evanston, IL, USA.,Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Swathi Kiran
- Center for the Neurobiology of Language Recovery, Northwestern University, Evanston, IL, USA.,Department of Speech, Language, and Hearing, College of Health & Rehabilitation, Boston University, Boston, MA, USA
| | - Brenda Rapp
- Center for the Neurobiology of Language Recovery, Northwestern University, Evanston, IL, USA.,Department of Cognitive Science, Krieger School of Arts & Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Cynthia Thompson
- Center for the Neurobiology of Language Recovery, Northwestern University, Evanston, IL, USA.,Department of Communication Sciences and Disorders, School of Communication, Northwestern University, Evanston, IL, USA.,Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Richard Zinbarg
- Center for the Neurobiology of Language Recovery, Northwestern University, Evanston, IL, USA.,Department of Psychology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA.,The Family Institute at Northwestern University, Evanston, IL, USA
| | - Todd Parrish
- Center for the Neurobiology of Language Recovery, Northwestern University, Evanston, IL, USA.,Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
30
|
Sahib AK, Loureiro JRA, Vasavada MM, Kubicki A, Joshi SH, Wang K, Woods RP, Congdon E, Wang DJJ, Boucher ML, Espinoza R, Narr KL. Single and repeated ketamine treatment induces perfusion changes in sensory and limbic networks in major depressive disorder. Eur Neuropsychopharmacol 2020; 33:89-100. [PMID: 32061453 PMCID: PMC8869841 DOI: 10.1016/j.euroneuro.2020.01.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/10/2020] [Accepted: 01/26/2020] [Indexed: 12/15/2022]
Abstract
Ketamine infusion therapy can produce fast-acting antidepressant effects in patients with major depressive disorder (MDD). Yet, how single and repeated ketamine treatment induces brain systems-level neuroplasticity underlying symptom improvement is unknown. Advanced multiband imaging (MB) pseudo-continuous arterial spin labeling (pCASL) perfusion MRI data was acquired from patients with treatment resistant depression (TRD) (N = 22, mean age=35.2 ± 9.95 SD, 27% female) at baseline, and 24 h after receiving single, and four subanesthetic (0.5 mg/kg) intravenous ketamine infusions. Changes in global and regional CBF were compared across time points, and relationships with overall mood, anhedonia and apathy were examined. Comparisons between patients at baseline and controls (N = 18, mean age=36.11 ± 14.5 SD, 57% female) established normalization of treatment effects. Results showed increased regional CBF in the cingulate and primary and higher-order visual association regions after first ketamine treatment. Baseline CBF in the fusiform, and acute changes in CBF in visual areas were related to symptom improvement after single and repeated ketamine treatment, respectively. In contrast, after serial infusion therapy, decreases in regional CBF were observed in the bilateral hippocampus and right insula with ketamine treatment. Findings demonstrate that neurophysiological changes occurring with single and repeated ketamine treatment follow both a regional and temporal pattern including sensory and limbic regions. Initial changes are observed in the posterior cingulate and precuneus and primary and higher-order visual areas, which relate to clinical responses. However, repeated exposure to ketamine, though not relating to clinical outcome, appears to engage deeper limbic structures and insula. ClinicalTrials.gov: Biomarkers of Fast Acting Therapies in Major Depression, https://clinicaltrials.gov/ct2/show/NCT02165449, NCT02165449.
Collapse
Affiliation(s)
- Ashish K Sahib
- Department of Neurology, Ahamason-Lovelace Brain Mapping Center, United States
| | - Joana R A Loureiro
- Department of Neurology, Ahamason-Lovelace Brain Mapping Center, United States
| | - Megha M Vasavada
- Department of Neurology, Ahamason-Lovelace Brain Mapping Center, United States
| | - Antoni Kubicki
- Department of Neurology, Ahamason-Lovelace Brain Mapping Center, United States
| | - Shantanu H Joshi
- Department of Neurology, Ahamason-Lovelace Brain Mapping Center, United States
| | - Kai Wang
- Laboratory of FMRI Technology (LOFT), Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, United States
| | - Roger P Woods
- Department of Neurology, Ahamason-Lovelace Brain Mapping Center, United States
| | - Eliza Congdon
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, 635 Charles E Young Drive South Suite, Los Angeles, CA 90095-7334, United States
| | - Danny J J Wang
- Laboratory of FMRI Technology (LOFT), Stevens Neuroimaging and Informatics Institute, University of Southern California, Los Angeles, CA, United States
| | - Michael L Boucher
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, 635 Charles E Young Drive South Suite, Los Angeles, CA 90095-7334, United States
| | - Randall Espinoza
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, 635 Charles E Young Drive South Suite, Los Angeles, CA 90095-7334, United States
| | - Katherine L Narr
- Department of Neurology, Ahamason-Lovelace Brain Mapping Center, United States; Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, 635 Charles E Young Drive South Suite, Los Angeles, CA 90095-7334, United States.
| |
Collapse
|
31
|
Yurgelun-Todd DA, Renshaw PF, Goldsmith P, Uz T, Macek TA. A randomized, placebo-controlled, phase 1 study to evaluate the effects of TAK-063 on ketamine-induced changes in fMRI BOLD signal in healthy subjects. Psychopharmacology (Berl) 2020; 237:317-328. [PMID: 31773211 PMCID: PMC7018803 DOI: 10.1007/s00213-019-05366-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 09/22/2019] [Indexed: 12/14/2022]
Abstract
RATIONALE Phosphodiesterase 10A inhibitor TAK-063 has shown effects that suggest efficacy in schizophrenia treatment. OBJECTIVE This randomized, double-blind, placebo-controlled, incomplete-crossover study investigated effects of single oral administration of TAK-063 on ketamine-induced changes in blood oxygen level-dependent (BOLD) signal in healthy males. METHODS Healthy men aged 18 to 45 years with normal magnetic resonance imaging (MRI) scans and electroencephalogram measurements at screening were eligible. Each subject was randomized to one of nine treatment schedules: all subjects received placebo and two of three doses of TAK-063 followed by ketamine. The primary endpoint was ketamine-induced brain activity in select regions of the brain during resting state. Secondary endpoints included pharmacokinetic parameters of TAK-063, proportion of subjects with treatment-emergent adverse events (AEs), and percentage of subjects meeting criteria for abnormal safety laboratory tests and vital sign measurements. RESULTS The study comprised 27 subjects. Prior to ketamine infusion, TAK-063 exerted region-specific effects on resting state functional MRI (fMRI) BOLD signal. After ketamine administration, TAK-063 reduced the Cohen's effect size for resting-state fMRI BOLD signal in key brain regions examined, and exerted similar effects on BOLD signal during the working memory task across all doses. TAK-063 was safe and well tolerated. CONCLUSIONS Our results are consistent with non-clinical studies of ketamine and TAK-063 and clinical studies of ketamine and risperidone. It is unknown whether these data are predictive of potential antipsychotic efficacy, and further analyses are required.
Collapse
Affiliation(s)
| | - Perry F Renshaw
- The Brain Institute, University of Utah, 383 Colorow Drive, Salt Lake City, UT, 84108, USA
| | - Paul Goldsmith
- Takeda Development Center Europe, Ltd., 61 Aldwych, London, WC2B 4AE, UK
| | - Tolga Uz
- Takeda Development Center Americas, Inc., One Takeda Parkway, Deerfield, IL, 60015, USA
| | - Thomas A Macek
- Takeda Development Center Americas, Inc., One Takeda Parkway, Deerfield, IL, 60015, USA
| |
Collapse
|
32
|
Forsyth A, McMillan R, Campbell D, Malpas G, Maxwell E, Sleigh J, Dukart J, Hipp J, Muthukumaraswamy SD. Modulation of simultaneously collected hemodynamic and electrophysiological functional connectivity by ketamine and midazolam. Hum Brain Mapp 2019; 41:1472-1494. [PMID: 31808268 PMCID: PMC7267972 DOI: 10.1002/hbm.24889] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 11/06/2019] [Accepted: 11/26/2019] [Indexed: 12/21/2022] Open
Abstract
The pharmacological modulation of functional connectivity in the brain may underlie therapeutic efficacy for several neurological and psychiatric disorders. Functional magnetic resonance imaging (fMRI) provides a noninvasive method of assessing this modulation, however, the indirect nature of the blood‐oxygen level dependent signal restricts the discrimination of neural from physiological contributions. Here we followed two approaches to assess the validity of fMRI functional connectivity in developing drug biomarkers, using simultaneous electroencephalography (EEG)/fMRI in a placebo‐controlled, three‐way crossover design with ketamine and midazolam. First, we compared seven different preprocessing pipelines to determine their impact on the connectivity of common resting‐state networks. Independent components analysis (ICA)‐denoising resulted in stronger reductions in connectivity after ketamine, and weaker increases after midazolam, than pipelines employing physiological noise modelling or averaged signals from cerebrospinal fluid or white matter. This suggests that pipeline decisions should reflect a drug's unique noise structure, and if this is unknown then accepting possible signal loss when choosing extensive ICA denoising pipelines could engender more confidence in the remaining results. We then compared the temporal correlation structure of fMRI to that derived from two connectivity metrics of EEG, which provides a direct measure of neural activity. While electrophysiological estimates based on the power envelope were more closely aligned to BOLD signal connectivity than those based on phase consistency, no significant relationship between the change in electrophysiological and hemodynamic correlation structures was found, implying caution should be used when making cross‐modal comparisons of pharmacologically‐modulated functional connectivity.
Collapse
Affiliation(s)
- Anna Forsyth
- School of Pharmacy, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Rebecca McMillan
- School of Pharmacy, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Doug Campbell
- Department of Anaesthesiology, Auckland District Health Board, Auckland, New Zealand
| | - Gemma Malpas
- Department of Anaesthesiology, Auckland District Health Board, Auckland, New Zealand
| | - Elizabeth Maxwell
- Department of Anaesthesiology, Auckland District Health Board, Auckland, New Zealand
| | - Jamie Sleigh
- Department of Anaesthesiology Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Juergen Dukart
- Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany.,Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jörg Hipp
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Suresh D Muthukumaraswamy
- School of Pharmacy, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
33
|
Duman RS, Shinohara R, Fogaça MV, Hare B. Neurobiology of rapid-acting antidepressants: convergent effects on GluA1-synaptic function. Mol Psychiatry 2019; 24:1816-1832. [PMID: 30894661 PMCID: PMC6754322 DOI: 10.1038/s41380-019-0400-x] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/22/2019] [Accepted: 03/05/2019] [Indexed: 11/09/2022]
Abstract
Efforts to develop efficacious antidepressant agents with novel mechanisms have been largely unsuccessful since the 1950's until the discovery of ketamine, an N-methyl-D-aspartate (NMDA) receptor antagonist that produces rapid and sustained antidepressant actions even in treatment-resistant patients. This finding has ushered in a new era for the development of novel rapid-acting antidepressants that act at the NMDA receptor complex, but without dissociative and psychotomimetic side effects of ketamine. Here, we review the current state of rapid-acting antidepressant drug development, including NMDA channel blockers, glycine site agents, and allosteric modulators, as well as ketamine stereoisomers and metabolites. In addition, we focus on the neurobiological mechanisms underlying the actions of these diverse agents and discuss evidence of convergent mechanisms including increased brain-derived neurotrophic factor signaling, increased synthesis of synaptic proteins, and most notably increased GluR1 and synaptic connectivity in the medial prefrontal cortex. These convergent mechanisms provide insight for potential additional novel targets for drug development (e.g., agents that increase synaptic protein synthesis and plasticity). Importantly, the convergent effects on synapse formation and plasticity also reverse the well-documented neuronal and synaptic deficits associated with stress and depression, and thereby target the underlying pathophysiology of major depressive disorder.
Collapse
Affiliation(s)
- Ronald S Duman
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
| | - Ryota Shinohara
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Manoela V Fogaça
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Brendan Hare
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
34
|
Amidfar M, Woelfer M, Réus GZ, Quevedo J, Walter M, Kim YK. The role of NMDA receptor in neurobiology and treatment of major depressive disorder: Evidence from translational research. Prog Neuropsychopharmacol Biol Psychiatry 2019; 94:109668. [PMID: 31207274 DOI: 10.1016/j.pnpbp.2019.109668] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 05/24/2019] [Accepted: 06/11/2019] [Indexed: 12/16/2022]
Abstract
There is accumulating evidence demonstrating that dysfunction of glutamatergic neurotransmission, particularly via N-methyl-d-aspartate (NMDA) receptors, is involved in the pathophysiology of major depressive disorder (MDD). Several studies have revealed an altered expression of NMDA receptor subtypes and impaired NMDA receptor-mediated intracellular signaling pathways in brain circuits of patients with MDD. Clinical studies have demonstrated that NMDA receptor antagonists, particularly ketamine, have rapid antidepressant effects in treatment-resistant depression, however, neurobiological mechanisms are not completely understood. Growing body of evidence suggest that signal transduction pathways involved in synaptic plasticity play critical role in molecular mechanisms underlying rapidly acting antidepressant properties of ketamine and other NMDAR antagonists in MDD. Discovering the molecular mechanisms underlying the unique antidepressant actions of ketamine will facilitate the development of novel fast acting antidepressants which lack undesirable effects of ketamine. This review provides a critical examination of the NMDA receptor involvement in the neurobiology of MDD including analyses of alterations in NMDA receptor subtypes and their interactive signaling cascades revealed by postmortem studies. Furthermore, to elucidate mechanisms underlying rapid-acting antidepressant properties of NMDA receptor antagonists we discussed their effects on the neuroplasticity, mostly based on signaling systems involved in synaptic plasticity of mood-related neurocircuitries.
Collapse
Affiliation(s)
| | - Marie Woelfer
- Clinical Affective Neuroimaging Laboratory, University Magdeburg, Germany; New Jersey Institute of Technology, Newark, NJ, USA
| | - Gislaine Z Réus
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil
| | - João Quevedo
- Translational Psychiatry Laboratory, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, Brazil; Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA; Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Martin Walter
- Clinical Affective Neuroimaging Laboratory, University Magdeburg, Germany; Department of Psychiatry, University Tuebingen, Germany
| | - Yong-Ku Kim
- Department of Psychiatry, College of Medicine, Korea University, Seoul, South Korea
| |
Collapse
|
35
|
Krajcovic B, Fajnerova I, Horacek J, Kelemen E, Kubik S, Svoboda J, Stuchlik A. Neural and neuronal discoordination in schizophrenia: From ensembles through networks to symptoms. Acta Physiol (Oxf) 2019; 226:e13282. [PMID: 31002202 DOI: 10.1111/apha.13282] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/27/2019] [Accepted: 04/12/2019] [Indexed: 12/22/2022]
Abstract
Despite the substantial knowledge accumulated by past research, the exact mechanisms of the pathogenesis of schizophrenia and causal treatments still remain unclear. Deficits of cognition and information processing in schizophrenia are today often viewed as the primary and core symptoms of this devastating disorder. These deficits likely result from disruptions in the coordination of neuronal and neural activity. The aim of this review is to bring together convergent evidence of discoordinated brain circuits in schizophrenia at multiple levels of resolution, ranging from principal cells and interneurons, neuronal ensembles and local circuits, to large-scale brain networks. We show how these aberrations could underlie deficits in cognitive control and other higher order cognitive-behavioural functions. Converging evidence from both animal models and patients with schizophrenia is presented in an effort to gain insight into common features of deficits in the brain information processing in this disorder, marked by disruption of several neurotransmitter and signalling systems and severe behavioural outcomes.
Collapse
Affiliation(s)
- Branislav Krajcovic
- Department of Neurophysiology of Memory Institute of Physiology of the Czech Academy of Sciences Prague Czech Republic
- Third Faculty of Medicine Charles University Prague Czech Republic
| | - Iveta Fajnerova
- Department of Neurophysiology of Memory Institute of Physiology of the Czech Academy of Sciences Prague Czech Republic
- Research Programme 3 - Applied Neurosciences and Brain Imaging National Institute of Mental Health Klecany Czech Republic
| | - Jiri Horacek
- Third Faculty of Medicine Charles University Prague Czech Republic
- Research Programme 3 - Applied Neurosciences and Brain Imaging National Institute of Mental Health Klecany Czech Republic
| | - Eduard Kelemen
- Research Programme 1 - Experimental Neurobiology National Institute of Mental Health Klecany Czech Republic
| | - Stepan Kubik
- Department of Neurophysiology of Memory Institute of Physiology of the Czech Academy of Sciences Prague Czech Republic
| | - Jan Svoboda
- Department of Neurophysiology of Memory Institute of Physiology of the Czech Academy of Sciences Prague Czech Republic
| | - Ales Stuchlik
- Department of Neurophysiology of Memory Institute of Physiology of the Czech Academy of Sciences Prague Czech Republic
| |
Collapse
|
36
|
Vlerick L, Peremans K, Dockx R, Audenaert K, Baeken C, Saunders JH, Polis I. The long-term effects of single and repeated subanaesthetic ketamine administration on regional cerebral blood flow in healthy dogs measured with 99mTc-HMPAO SPECT. Psychiatry Res Neuroimaging 2019; 285:18-24. [PMID: 30716686 DOI: 10.1016/j.pscychresns.2019.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 01/15/2019] [Accepted: 01/15/2019] [Indexed: 12/13/2022]
Abstract
Subanaesthetic ketamine has recently been established as an effective and rapid treatment for major depressive disorder showing antidepressant effects for up to 1 week on average. The use of repeated ketamine infusions has been put forward to augment and to prolong the antidepressant response and increase the remission rates. The underlying neurobiological mechanisms responsible for ketamine's antidepressant effects remain unclear. Nevertheless, it has been shown, both in dogs and humans, that ketamine can alter neuronal perfusion and therefore neuronal function in brain regions involved in psychiatric and behavioural disorders. Consequently, the aim of the current placebo controlled study was to assess the long-term effects on cerebral perfusion of single and repeated subanaesthetic ketamine infusions in dogs. Twelve healthy, laboratory dogs were scanned at six different time points following single and repeated ketamine administration, using Single Photon Emission Computed Tomography with the radiotracer 99mTc-hexamethylpropylene amine oxime. We hypothesised that repeated infusions could lead to more prolonged perfusion alterations in brain regions critical for behaviour regulation. We found that repeated subanaesthetic ketamine administration did not result in more prolonged cerebral perfusion alterations compared to a single ketamine administration.
Collapse
Affiliation(s)
- Lise Vlerick
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, East Flanders, Belgium.
| | - Kathelijne Peremans
- Department of Veterinary Medical Imaging and Small Animal Orthopaedics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, East Flanders, Belgium
| | - Robrecht Dockx
- Ghent Experimental Psychiatry (GHEP) lab, Department of Psychiatry and Medical Psychology, Ghent University, Ghent, East Flanders, Belgium; Department of Psychiatry, University Hospital (UZBrussel), Brussels, Belgium
| | - Kurt Audenaert
- Ghent Experimental Psychiatry (GHEP) lab, Department of Psychiatry and Medical Psychology, Ghent University, Ghent, East Flanders, Belgium; Department of Psychiatry, University Hospital (UZBrussel), Brussels, Belgium
| | - Chris Baeken
- Ghent Experimental Psychiatry (GHEP) lab, Department of Psychiatry and Medical Psychology, Ghent University, Ghent, East Flanders, Belgium; Department of Psychiatry, University Hospital (UZBrussel), Brussels, Belgium
| | - Jimmy H Saunders
- Department of Veterinary Medical Imaging and Small Animal Orthopaedics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, East Flanders, Belgium
| | - Ingeborgh Polis
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, East Flanders, Belgium
| |
Collapse
|
37
|
Hahn A, Lanzenberger R, Kasper S. Making Sense of Connectivity. Int J Neuropsychopharmacol 2019; 22:194-207. [PMID: 30544240 PMCID: PMC6403091 DOI: 10.1093/ijnp/pyy100] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 11/07/2018] [Accepted: 12/11/2018] [Indexed: 02/07/2023] Open
Abstract
In addition to the assessment of local alterations of specific brain regions, the investigation of entire networks with in vivo neuroimaging techniques has gained increasing attention. In general, connectivity analysis refers to the investigation of links between brain regions, with the aim to characterize their interactions and information transfer. These may represent or relate to different physiological characteristics (structural, functional, or metabolic information) and can be calculated across different levels of granularity (2 regions vs whole brain). In this article, we provide an overview of different connectivity analysis approaches with interpretations and limitations as well as examples in pharmacological imaging and clinical applications. Structural connectivity obtained from diffusion MRI enables the reconstruction of neuronal fiber tracts. These physical links represent major constraints of functional connections, which are in turn defined as correlations between signal time courses. In addition, molecular connectivity approaches based on PET imaging enable the assessment of interregional associations of metabolic demands and neurotransmitter systems. Application of these approaches in clinical investigations has demonstrated novel alterations in various neurological and psychiatric disorders on a network level. Future work should aim for the combined assessment of multiple imaging modalities and to establish robust biomarkers for clinical use. These advancements will further improve the biological interpretation of connectivity metrics and networks of the human brain.
Collapse
Affiliation(s)
- Andreas Hahn
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| | - Siegfried Kasper
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Austria
| |
Collapse
|
38
|
Fleming LM, Javitt DC, Carter CS, Kantrowitz JT, Girgis RR, Kegeles LS, Ragland JD, Maddock RJ, Lesh TA, Tanase C, Robinson J, Potter WZ, Carlson M, Wall MM, Choo TH, Grinband J, Lieberman J, Krystal JH, Corlett PR. A multicenter study of ketamine effects on functional connectivity: Large scale network relationships, hubs and symptom mechanisms. NEUROIMAGE-CLINICAL 2019; 22:101739. [PMID: 30852397 PMCID: PMC6411494 DOI: 10.1016/j.nicl.2019.101739] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 02/24/2019] [Accepted: 02/27/2019] [Indexed: 12/14/2022]
Abstract
Ketamine is an uncompetitive N-methyl-d-aspartate (NMDA) glutamate receptor antagonist. It induces effects in healthy individuals that mimic symptoms associated with schizophrenia. We sought to root these experiences in altered brain function, specifically aberrant resting state functional connectivity (rsfMRI). In the present study, we acquired rsfMRI data under ketamine and placebo in a between-subjects design and analyzed seed-based measures of rsfMRI using large-scale networks, dorsolateral prefrontal cortex (DLPFC) and sub-nuclei of the thalamus. We found ketamine-induced alterations in rsfMRI connectivity similar to those seen in patients with schizophrenia, some changes that may be more comparable to early stages of schizophrenia, and other connectivity signatures seen in patients that ketamine did not recreate. We do not find any circuits from our regions of interest that correlates with positive symptoms of schizophrenia in our sample, although we find that DLPFC connectivity with ACC does correlate with a mood measure. These results provide support for ketamine's use as a model of certain biomarkers of schizophrenia, particularly for early or at-risk patients. Ketamine altered connectivity in default mode network, thalamus and DLPFC at rest. Similar patterns of altered connectivity are seen with ketamine as in psychotic patients. Mood symptoms correlated with DLPFC connectivity with ACC and frontal orbital cortex.
Collapse
Affiliation(s)
- Leah M Fleming
- Department of Psychiatry, Yale University, New Haven, CT, United States of America; Interdepartmental Neuroscience Program, Yale University, New Haven, CT, United States of America
| | - Daniel C Javitt
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY, United States of America; Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, NY, United States of America
| | - Cameron S Carter
- Department of Psychiatry, University of California, Davis, United States of America
| | - Joshua T Kantrowitz
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY, United States of America; Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, NY, United States of America
| | - Ragy R Girgis
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY, United States of America
| | - Lawrence S Kegeles
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY, United States of America
| | - John D Ragland
- Department of Psychiatry, University of California, Davis, United States of America
| | - Richard J Maddock
- Department of Psychiatry, University of California, Davis, United States of America
| | - Tyler A Lesh
- Department of Psychiatry, University of California, Davis, United States of America
| | - Costin Tanase
- Department of Psychiatry, University of California, Davis, United States of America
| | - James Robinson
- Nathan Kline Institute for Psychiatric Research, Orangeburg, New York, NY, United States of America
| | - William Z Potter
- National Institute of Mental Health, Rockville, MD, United States of America
| | - Marlene Carlson
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY, United States of America
| | - Melanie M Wall
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY, United States of America; National Institute of Mental Health, Rockville, MD, United States of America
| | - Tse-Hwei Choo
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY, United States of America
| | - Jack Grinband
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY, United States of America
| | - Jeffrey Lieberman
- Department of Psychiatry, New York State Psychiatric Institute, Columbia University Medical Center, New York, NY, United States of America
| | - John H Krystal
- Department of Psychiatry, Yale University, New Haven, CT, United States of America
| | - Philip R Corlett
- Department of Psychiatry, Yale University, New Haven, CT, United States of America.
| |
Collapse
|
39
|
McMillan R, Forsyth A, Campbell D, Malpas G, Maxwell E, Dukart J, Hipp JF, Muthukumaraswamy S. Temporal dynamics of the pharmacological MRI response to subanaesthetic ketamine in healthy volunteers: A simultaneous EEG/fMRI study. J Psychopharmacol 2019; 33:219-229. [PMID: 30663520 DOI: 10.1177/0269881118822263] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Pharmacological magnetic resonance imaging has been used to investigate the neural effects of subanaesthetic ketamine in healthy volunteers. However, the effect of ketamine has been modelled with a single time course and without consideration of physiological noise. AIMS This study aimed to investigate ketamine-induced alterations in resting neural activity using conventional pharmacological magnetic resonance imaging analysis techniques with physiological noise correction, and a novel analysis utilising simultaneously recorded electroencephalography data. METHODS Simultaneous electroencephalography/functional magnetic resonance imaging and physiological data were collected from 30 healthy male participants before and during a subanaesthetic intravenous ketamine infusion. RESULTS Consistent with previous literature, we show widespread cortical blood-oxygen-level dependent signal increases and decreased blood-oxygen-level dependent signals in the subgenual anterior cingulate cortex following ketamine. However, the latter effect was attenuated by the inclusion of motion regressors and physiological correction in the model. In a novel analysis, we modelled the pharmacological magnetic resonance imaging response with the power time series of seven electroencephalography frequency bands. This showed evidence for distinct temporal time courses of neural responses to ketamine. No electroencephalography power time series correlated with decreased blood-oxygen-level dependent signal in the subgenual anterior cingulate cortex. CONCLUSIONS We suggest the decrease in blood-oxygen-level dependent signals in the subgenual anterior cingulate cortex typically seen in the literature is the result of physiological noise, in particular cardiac pulsatility. Furthermore, modelling the pharmacological magnetic resonance imaging response with a single temporal model does not completely capture the full spectrum of neuronal dynamics. The use of electroencephalography regressors to model the response can increase confidence that the pharmacological magnetic resonance imaging is directly related to underlying neural activity.
Collapse
Affiliation(s)
- Rebecca McMillan
- 1 School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Anna Forsyth
- 1 School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Doug Campbell
- 2 Department of Anaesthesiology, Auckland District Health Board, Auckland, New Zealand
| | - Gemma Malpas
- 2 Department of Anaesthesiology, Auckland District Health Board, Auckland, New Zealand
| | - Elizabeth Maxwell
- 2 Department of Anaesthesiology, Auckland District Health Board, Auckland, New Zealand
| | - Juergen Dukart
- 3 F. Hoffmann-La Roche, Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland.,4 Institute of Neuroscience and Medicine, Brain & Behaviour (INM-7), Research Centre Jülich, Jülich, Germany.,5 Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Joerg F Hipp
- 3 F. Hoffmann-La Roche, Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland
| | - Suresh Muthukumaraswamy
- 1 School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
40
|
Test-retest reliability of task-based and resting-state blood oxygen level dependence and cerebral blood flow measures. PLoS One 2018; 13:e0206583. [PMID: 30408072 PMCID: PMC6224062 DOI: 10.1371/journal.pone.0206583] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 10/16/2018] [Indexed: 11/20/2022] Open
Abstract
Despite their wide-spread use, only limited information is available on the comparative test-retest reliability of task-based functional and resting state magnetic resonance imaging measures of blood oxygen level dependence (tb-fMRI and rs-fMRI) and cerebral blood flow (CBF) using arterial spin labeling. This information is critical to designing properly powered longitudinal studies. Here we comprehensively quantified and compared the test-retest reliability and reproducibility performance of 8 commonly applied fMRI tasks, 6 rs-fMRI metrics and CBF in 30 healthy volunteers. We find large variability in test-retest reliability performance across the different tb-fMRI paradigms and rs-fMRI metrics, ranging from poor to excellent. A larger extent of activation in tb-fMRI is linked to higher between-subject reliability of the respective task suggesting that differences in the amount of activation may be used as a first reliability estimate of novel tb-fMRI paradigms. For rs-fMRI, a good reliability of local activity estimates is paralleled by poor performance of global connectivity metrics. Evaluated CBF measures provide in general a good to excellent test-reliability matching or surpassing the best performing tb-fMRI and rs-fMRI metrics. This comprehensive effort allows for direct comparisons of test-retest reliability between the evaluated MRI domains and measures to aid the design of future tb-fMRI, rs-fMRI and CBF studies.
Collapse
|
41
|
Haaf M, Leicht G, Curic S, Mulert C. Glutamatergic Deficits in Schizophrenia - Biomarkers and Pharmacological Interventions within the Ketamine Model. Curr Pharm Biotechnol 2018; 19:293-307. [PMID: 29929462 PMCID: PMC6142413 DOI: 10.2174/1389201019666180620112528] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/12/2018] [Accepted: 05/26/2018] [Indexed: 11/30/2022]
Abstract
Background: The observation that N-methyl-D-aspartate glutamate receptor (NMDAR) antagonists such as ketamine transiently induce schizophrenia-like positive, negative and cognitive symptoms has led to a paradigm shift from dopaminergic to glutamatergic dysfunction in pharmacological models of schizophrenia. NMDAR hypofunction can explain many schizophrenia symptoms directly due to excitatory-to-inhibitory (E/I) imbalance, but also dopaminergic dysfunction itself. However, so far no new drug targeting the NMDAR has been successfully approved. In the search for possible biomarkers it is interesting that ketamine-induced psychopathological changes in healthy participants were accompanied by altered electro-(EEG), magnetoencephalographic (MEG) and functional magnetic resonance imaging (fMRI) signals. Methods: We systematically searched PubMed/Medline and Web of Knowledge databases (January 2006 to July 2017) to identify EEG/MEG and fMRI studies of the ketamine model of schizophrenia with human subjects. The search strategy identified 209 citations of which 46 articles met specified eligibility criteria. Results: In EEG/MEG studies, ketamine induced changes of event-related potentials, such as the P300 potential and the mismatch negativity, similar to alterations observed in schizophrenia patients. In fMRI studies, alterations of activation were observed in different brain regions, most prominently within the anterior cingulate cortex and limbic structures as well as task-relevant brain regions. These alterations were accompanied by changes in functional connectivity, indicating a balance shift of the underlying brain networks. Pharmacological treatments did alter ketamine-induced changes in EEG/MEG and fMRI studies to different extents. Conclusion: This review highlights the potential applicability of the ketamine model for schizophrenia drug development by offering the possibility to assess the effect of pharmacological agents on schizophrenia-like symptoms and to find relevant neurophysiological and neuroimaging biomarkers.
Collapse
Affiliation(s)
- Moritz Haaf
- Psychiatry Neuroimaging Branch (PNB), Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gregor Leicht
- Psychiatry Neuroimaging Branch (PNB), Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stjepan Curic
- Psychiatry Neuroimaging Branch (PNB), Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoph Mulert
- Psychiatry Neuroimaging Branch (PNB), Department of Psychiatry and Psychotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Psychiatry and Psychotherapy, UKGM, Justus-Liebig University Giessen, Giessen, Germany
| |
Collapse
|
42
|
Evans JW, Szczepanik J, Brutsché N, Park LT, Nugent AC, Zarate CA. Default Mode Connectivity in Major Depressive Disorder Measured Up to 10 Days After Ketamine Administration. Biol Psychiatry 2018; 84:582-590. [PMID: 29580569 PMCID: PMC6093808 DOI: 10.1016/j.biopsych.2018.01.027] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/05/2018] [Accepted: 01/23/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND The symptoms of major depressive disorder (MDD) are rapidly alleviated by administration of a single dose of the glutamatergic modulator ketamine. However, few studies have investigated the potential sustained neural effects of this agent beyond immediate infusion. This study used functional magnetic resonance imaging to examine the effect of a single ketamine infusion on the resting state default mode network (DMN) at 2 and 10 days after a single ketamine infusion in unmedicated subjects with MDD as well as healthy control subjects (HCs). METHODS Data were drawn from a double-blind, placebo-controlled crossover study of 58 participants (33 with MDD and 25 HCs) who received an intravenous infusion of either ketamine hydrochloride (0.5 mg/kg) or placebo on 2 separate test days spaced 2 weeks apart. Eight minutes of functional magnetic resonance imaging resting state data was acquired at baseline and at about 2 and 10 days after both infusions. The DMN was defined using seed-based correlation and was compared across groups and scans. RESULTS In subjects with MDD, connectivity between the insula and the DMN was normalized compared with HCs 2 days postketamine infusion. This change was reversed after 10 days and did not appear in either of the placebo scans. Group-specific connectivity differences in drug response were observed, most notably in the insula in subjects with MDD and in the thalamus in HCs. CONCLUSIONS Connectivity changes in the insula in subjects with MDD suggest that ketamine may normalize the interaction between the DMN and salience networks, supporting the triple network dysfunction model of MDD.
Collapse
Affiliation(s)
- Jennifer W Evans
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland.
| | - Joanna Szczepanik
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Nancy Brutsché
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Lawrence T Park
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Allison C Nugent
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
43
|
Ketamine and pharmacological imaging: use of functional magnetic resonance imaging to evaluate mechanisms of action. Behav Pharmacol 2018; 28:610-622. [PMID: 29049083 DOI: 10.1097/fbp.0000000000000354] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Ketamine has been used as a pharmacological model for schizophrenia as subanesthetic infusions have been shown to produce temporary schizophrenia-like symptoms in healthy humans. More recently, ketamine has emerged as a potential treatment for multiple psychiatric disorders, including treatment-resistant depression and suicidal ideation. However, the mechanisms underlying both the psychotomimetic and the therapeutic effects of ketamine remain poorly understood. This review provides an overview of what is known of the neural mechanisms underlying the effects of ketamine and details what functional MRI studies have yielded at a systems level focused on brain circuitry. Multiple analytic approaches show that ketamine exerts robust and consistent effects at the whole-brain level. These effects are highly conserved across human and nonhuman primates, validating the use of nonhuman primate models for further investigations with ketamine. Regional analysis of brain functional connectivity suggests that the therapeutic potential of ketamine may be derived from a strengthening of executive control circuitry, making it an intriguing candidate for the treatment of drug abuse. There are still important questions about the mechanism of action and the therapeutic potential of ketamine that can be addressed using appropriate functional neuroimaging techniques.
Collapse
|
44
|
Temporally dissociable effects of ketamine on neuronal discharge and gamma oscillations in rat thalamo-cortical networks. Neuropharmacology 2018; 137:13-23. [DOI: 10.1016/j.neuropharm.2018.04.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/19/2018] [Accepted: 04/21/2018] [Indexed: 12/28/2022]
|
45
|
Mehta MA, Schmechtig A, Kotoula V, McColm J, Jackson K, Brittain C, Tauscher-Wisniewski S, Kinon BJ, Morrison PD, Pollak T, Mant T, Williams SCR, Schwarz AJ. Group II metabotropic glutamate receptor agonist prodrugs LY2979165 and LY2140023 attenuate the functional imaging response to ketamine in healthy subjects. Psychopharmacology (Berl) 2018; 235:1875-1886. [PMID: 29564482 DOI: 10.1007/s00213-018-4877-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 03/08/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND Aberrant glutamate neurotransmission, and in particular dysfunction of the N-methyl-D-aspartate receptor (NMDAR), has been implicated in psychiatric disorders and represents a novel therapeutic target. Low-dose administration of the NMDA antagonist ketamine in healthy volunteers elicits a strong blood oxygenation level dependent (BOLD) imaging signal that can be attenuated by pretreatment with single, therapeutically effective doses of marketed medicines interacting with the glutamate system. OBJECTIVE To test the attenuation of the ketamine-induced BOLD signal by pretreatment with either a metabotropic glutamate receptor (mGluR) 2/3 or a mGluR2 agonist in healthy volunteers METHODS: We used a ketamine challenge pharmacological magnetic resonance imaging (phMRI) paradigm to assess the modulatory effects of single acute doses of LY2140023 (pomaglumetad methionil), the methionine prodrug of the mGluR2/3 agonist LY404039 (10, 40, and 160 mg; N = 16 subjects) and of LY2979165, and the alanine prodrug of the selective orthosteric mGluR2 agonist 2812223 (20 and 60 mg; N = 16 subjects). RESULTS A reduction in the ketamine-evoked BOLD phMRI signal relative to placebo was observed at the highest doses tested of both LY2140023 and LY2979165. A relationship was observed between reduction of the BOLD signal and increasing plasma levels of 2812223 in the LY2979165 cohort. CONCLUSIONS These results identify pharmacologically active doses of the group II mGluR agonist prodrugs LY2140023 and LY2979165 in humans. They also extend the classes of compounds that have been experimentally shown to reverse the ketamine-evoked phMRI signal in humans, further supporting the use of this method as a neuroimaging biomarker for assessing functional effects.
Collapse
Affiliation(s)
- Mitul A Mehta
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK.
| | - Anne Schmechtig
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK
| | - Vasileia Kotoula
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK
| | - Juliet McColm
- Eli Lilly and Company, Sunninghill Road, Windlesham, Surrey, UK
| | | | - Claire Brittain
- Eli Lilly and Company, Sunninghill Road, Windlesham, Surrey, UK
| | | | | | - Paul D Morrison
- Psychosis Studies Department, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Thomas Pollak
- Psychosis Studies Department, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | | | - Steven C R Williams
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK
| | | |
Collapse
|
46
|
Bruinsma TJ, Sarma VV, Oh Y, Jang DP, Chang SY, Worrell GA, Lowe VJ, Jo HJ, Min HK. The Relationship Between Dopamine Neurotransmitter Dynamics and the Blood-Oxygen-Level-Dependent (BOLD) Signal: A Review of Pharmacological Functional Magnetic Resonance Imaging. Front Neurosci 2018; 12:238. [PMID: 29692706 PMCID: PMC5902685 DOI: 10.3389/fnins.2018.00238] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 03/27/2018] [Indexed: 11/13/2022] Open
Abstract
Functional magnetic resonance imaging (fMRI) is widely used in investigations of normal cognition and brain disease and in various clinical applications. Pharmacological fMRI (pharma-fMRI) is a relatively new application, which is being used to elucidate the effects and mechanisms of pharmacological modulation of brain activity. Characterizing the effects of neuropharmacological agents on regional brain activity using fMRI is challenging because drugs modulate neuronal function in a wide variety of ways, including through receptor agonist, antagonist, and neurotransmitter reuptake blocker events. Here we review current knowledge on neurotransmitter-mediated blood-oxygen-level dependent (BOLD) fMRI mechanisms as well as recently updated methodologies aimed at more fully describing the effects of neuropharmacologic agents on the BOLD signal. We limit our discussion to dopaminergic signaling as a useful lens through which to analyze and interpret neurochemical-mediated changes in the hemodynamic BOLD response. We also discuss the need for future studies that use multi-modal approaches to expand the understanding and application of pharma-fMRI.
Collapse
Affiliation(s)
- Tyler J Bruinsma
- Department of Radiology, College of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Vidur V Sarma
- Department of Radiology, College of Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN, United States
| | - Yoonbae Oh
- Department of Biomedical Engineering, Hanyang University, Seoul, South Korea.,Department of Neurologic Surgery, College of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Dong Pyo Jang
- Department of Biomedical Engineering, Hanyang University, Seoul, South Korea
| | - Su-Youne Chang
- Department of Neurologic Surgery, College of Medicine, Mayo Clinic, Rochester, MN, United States.,Departments of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| | - Greg A Worrell
- Department of Neurology, College of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Val J Lowe
- Department of Radiology, College of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Hang Joon Jo
- Department of Neurologic Surgery, College of Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Neurology, College of Medicine, Mayo Clinic, Rochester, MN, United States
| | - Hoon-Ki Min
- Department of Radiology, College of Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Neurologic Surgery, College of Medicine, Mayo Clinic, Rochester, MN, United States.,Departments of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
47
|
Iven J, Biesiekierski JR, Zhao D, Deloose E, O'Daly OG, Depoortere I, Tack J, Van Oudenhove L. Intragastric quinine administration decreases hedonic eating in healthy women through peptide-mediated gut-brain signaling mechanisms. Nutr Neurosci 2018; 22:850-862. [PMID: 29607741 DOI: 10.1080/1028415x.2018.1457841] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Objectives: Intragastric bitter tastants may decrease appetite and food intake. We aimed to investigate the gut-brain signaling and brain mechanisms underlying these effects.Methods: Brain responses to intragastric quinine-hydrochloride (QHCl, 10 µmol/kg) or placebo infusion were recorded using functional magnetic resonance imaging in 15 healthy women. Appetite-related sensations, plasma levels of gastrointestinal hormones and hedonic food intake (ad libitum drink test) were assessed.Results: Lower octanoylated ghrelin (P<0.04), total ghrelin (P<0.01), and motilin (P<0.01) plasma levels were found after QHCl administration, along with lower prospective food consumption ratings (P<0.02) and hedonic food intake (P<0.05). QHCl increased neural activity in the hypothalamus and hedonic (anterior insula, putamen, caudate, pallidum, amygdala, anterior cingulate cortex, orbitofrontal cortex, midbrain) regions, but decreased activity in the homeostatic medulla (all pFWE-corrected<0.05). Differential brain responses to QHCl versus placebo covaried with subjective and hormonal responses and predicted differences in hedonic food intake.Discussion: Intragastric QHCl decreases prospective and actual food intake in healthy women by interfering with homeostatic and hedonic brain circuits in a ghrelin- and motilin-mediated fashion. These findings suggest a potential of bitter tastants to reduce appetite and food intake, through the gut-brain axis.
Collapse
Affiliation(s)
- Julie Iven
- Laboratory for Brain-Gut Axis Studies (LaBGAS), Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Jessica R Biesiekierski
- Laboratory for Brain-Gut Axis Studies (LaBGAS), Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Dongxing Zhao
- Laboratory for Brain-Gut Axis Studies (LaBGAS), Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Eveline Deloose
- Laboratory for Brain-Gut Axis Studies (LaBGAS), Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Owen G O'Daly
- Center for Neuroimaging Sciences, Psychology, and Neurosciences, Institute of Psychiatry, King's College London, London, UK
| | - Inge Depoortere
- Laboratory for Brain-Gut Axis Studies (LaBGAS), Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Jan Tack
- Laboratory for Brain-Gut Axis Studies (LaBGAS), Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Lukas Van Oudenhove
- Laboratory for Brain-Gut Axis Studies (LaBGAS), Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| |
Collapse
|
48
|
Tang H, Kukral D, Li YW, Fronheiser M, Malone H, Pena A, Pieschl R, Sidik K, Tobon G, Chow PL, Bristow LJ, Hayes W, Luo F. Mapping the central effects of (±)-ketamine and traxoprodil using pharmacological magnetic resonance imaging in awake rats. J Psychopharmacol 2018; 32:146-155. [PMID: 29378483 DOI: 10.1177/0269881117746901] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Major depressive disorder is a leading cause of disability globally. Improvements in the efficacy of antidepressant therapy are needed as a high proportion (>40%) of individuals with major depressive disorder fail to respond adequately to current treatments. The non-selective N-methyl-D-aspartate receptor channel blocker, (±)-ketamine, has been reported to produce a rapid and long-lasting antidepressant response in treatment-resistant major depressive disorder patients, which provides a unique opportunity for investigation of mechanisms that mediate its therapeutic effect. Efforts have also focused on the development of selective N-methyl-D-aspartate receptor subtype 2B antagonists which may retain antidepressant activity but have lower potential for dissociative/psychotomimetic effects. In the present study, we examined the central nervous system effects of acute, intravenous administration of (±)-ketamine or the N-methyl-D-aspartate receptor subtype 2B antagonist, traxoprodil, in awake rats using pharmacological magnetic resonance imaging. The study contained five treatment groups: vehicle, 3 mg/kg (±)-ketamine, and three doses of traxoprodil (0.3 mg/kg, 5 mg/kg, and 15 mg/kg). Non-linear model fitting was performed on the temporal hemodynamic pharmacological magnetic resonance imaging data to generate brain activation maps as well as regional responses based on blood oxygen level dependent signal changes for group analysis. Traxoprodil at 5 mg/kg and 15 mg/kg produced a dose-dependent pharmacological magnetic resonance imaging signal in rat forebrain regions with both doses achieving >80% N-methyl-D-aspartate receptor subtype 2B occupancy determined by ex vivo [3H]Ro 25-6981 binding. The middle dose of traxoprodil (5 mg/kg) generated region-specific activations in medial prefrontal cortex, ventral orbital cortex, and anterior cingulate cortex whereas the high dose (15 mg/kg) produced a widespread pharmacological magnetic resonance imaging response in both cortical and subcortical brain regions which was similar to that produced by (±)-ketamine (3 mg/kg, intravenous).
Collapse
Affiliation(s)
- Haiying Tang
- 1 Translational Sciences, Bristol-Myers Squibb, Lawrenceville, USA
| | - Daniel Kukral
- 1 Translational Sciences, Bristol-Myers Squibb, Lawrenceville, USA
| | - Yu-Wen Li
- 2 Discovery Biology, Bristol-Myers Squibb, Wallingford, USA
| | | | - Harold Malone
- 1 Translational Sciences, Bristol-Myers Squibb, Lawrenceville, USA
| | - Adrienne Pena
- 1 Translational Sciences, Bristol-Myers Squibb, Lawrenceville, USA
| | - Rick Pieschl
- 2 Discovery Biology, Bristol-Myers Squibb, Wallingford, USA
| | - Kurex Sidik
- 3 Global Biometrics Sciences, Bristol-Myers Squibb, Lawrenceville, USA
| | | | - Patrick L Chow
- 1 Translational Sciences, Bristol-Myers Squibb, Lawrenceville, USA
| | | | - Wendy Hayes
- 1 Translational Sciences, Bristol-Myers Squibb, Lawrenceville, USA
| | - Feng Luo
- 5 Translational Sciences, Bristol-Myers Squibb, Wallingford, USA
| |
Collapse
|
49
|
Carmichael O, Schwarz AJ, Chatham CH, Scott D, Turner JA, Upadhyay J, Coimbra A, Goodman JA, Baumgartner R, English BA, Apolzan JW, Shankapal P, Hawkins KR. The role of fMRI in drug development. Drug Discov Today 2018; 23:333-348. [PMID: 29154758 PMCID: PMC5931333 DOI: 10.1016/j.drudis.2017.11.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/19/2017] [Accepted: 11/13/2017] [Indexed: 12/17/2022]
Abstract
Functional magnetic resonance imaging (fMRI) has been known for over a decade to have the potential to greatly enhance the process of developing novel therapeutic drugs for prevalent health conditions. However, the use of fMRI in drug development continues to be relatively limited because of a variety of technical, biological, and strategic barriers that continue to limit progress. Here, we briefly review the roles that fMRI can have in the drug development process and the requirements it must meet to be useful in this setting. We then provide an update on our current understanding of the strengths and limitations of fMRI as a tool for drug developers and recommend activities to enhance its utility.
Collapse
Affiliation(s)
- Owen Carmichael
- Pennington Biomedical Research Center, Baton Rouge, LA, USA.
| | | | - Christopher H Chatham
- Translational Medicine Neuroscience and Biomarkers, Roche Innovation Center, Basel, Switzerland
| | | | - Jessica A Turner
- Psychology Department & Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | | | | | | | - Richard Baumgartner
- Biostatistics and Research Decision Sciences (BARDS), Merck & Co., Inc., Kenilworth, NJ, USA
| | | | - John W Apolzan
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | | | | |
Collapse
|
50
|
The motilin agonist erythromycin increases hunger by modulating homeostatic and hedonic brain circuits in healthy women: a randomized, placebo-controlled study. Sci Rep 2018; 8:1819. [PMID: 29379095 PMCID: PMC5789052 DOI: 10.1038/s41598-018-19444-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/02/2018] [Indexed: 02/08/2023] Open
Abstract
The motilin agonist, erythromycin, induces gastric phase III of the migrating motor complex, which in turn generates hunger peaks. To identify the brain mechanisms underlying these orexigenic effects, 14 healthy women participated in a randomized, placebo-controlled crossover study. Functional magnetic resonance brain images were acquired for 50 minutes interprandially. Intravenous infusion of erythromycin (40 mg) or saline started 10 minutes after the start of scanning. Blood samples (for glucose and hormone levels) and hunger ratings were collected at fixed timepoints. Thirteen volunteers completed the study, without any adverse events. Brain regions involved in homeostatic and hedonic control of appetite and food intake responded to erythromycin, including pregenual anterior cingulate cortex, anterior insula cortex, orbitofrontal cortex, amygdala, caudate, pallidum and putamen bilaterally, right accumbens, hypothalamus, and midbrain. Octanoylated ghrelin levels decreased, whereas both glucose and insulin increased after erythromycin. Hunger were higher after erythromycin, and these differences covaried with the brain response in most of the abovementioned regions. The motilin agonist erythromycin increases hunger by modulating neurocircuitry related to homeostatic and hedonic control of appetite and feeding. These results confirm recent behavioural findings identifying motilin as a key orexigenic hormone in humans, and identify the brain mechanisms underlying its effect.
Collapse
|