1
|
Agnorelli C, Spriggs M, Godfrey K, Sawicka G, Bohl B, Douglass H, Fagiolini A, Parastoo H, Carhart-Harris R, Nutt D, Erritzoe D. Neuroplasticity and psychedelics: A comprehensive examination of classic and non-classic compounds in pre and clinical models. Neurosci Biobehav Rev 2025; 172:106132. [PMID: 40185376 DOI: 10.1016/j.neubiorev.2025.106132] [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: 11/29/2024] [Revised: 03/24/2025] [Accepted: 03/29/2025] [Indexed: 04/07/2025]
Abstract
Neuroplasticity, the ability of the nervous system to adapt throughout an organism's lifespan, offers potential as both a biomarker and treatment target for neuropsychiatric conditions. Psychedelics, a burgeoning category of drugs, are increasingly prominent in psychiatric research, prompting inquiries into their mechanisms of action. Distinguishing themselves from traditional medications, psychedelics demonstrate rapid and enduring therapeutic effects after a single or few administrations, believed to stem from their neuroplasticity-enhancing properties. This review examines how classic psychedelics (e.g., LSD, psilocybin, N,N-DMT) and non-classic psychedelics (e.g., ketamine, MDMA) influence neuroplasticity. Drawing from preclinical and clinical studies, we explore the molecular, structural, and functional changes triggered by these agents. Animal studies suggest psychedelics induce heightened sensitivity of the nervous system to environmental stimuli (meta-plasticity), re-opening developmental windows for long-term structural changes (hyper-plasticity), with implications for mood and behavior. Translating these findings to humans faces challenges due to limitations in current imaging techniques. Nonetheless, promising new directions for human research are emerging, including the employment of novel positron-emission tomography (PET) radioligands, non-invasive brain stimulation methods, and multimodal approaches. By elucidating the interplay between psychedelics and neuroplasticity, this review informs the development of targeted interventions for neuropsychiatric disorders and advances understanding of psychedelics' therapeutic potential.
Collapse
Affiliation(s)
- Claudio Agnorelli
- Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, UK; Unit of Psychiatry, Department of Molecular and Developmental Medicine, University of Siena, Italy.
| | - Meg Spriggs
- Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, UK
| | - Kate Godfrey
- Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, UK
| | - Gabriela Sawicka
- Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, UK
| | - Bettina Bohl
- Department of Bioengineering, Imperial College of London, UK
| | - Hannah Douglass
- Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, UK
| | - Andrea Fagiolini
- Unit of Psychiatry, Department of Molecular and Developmental Medicine, University of Siena, Italy
| | | | - Robin Carhart-Harris
- Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, UK; Departments of Neurology and Psychiatry, Carhart-Harris Lab, University of California San Francisco, San Francisco, CA, USA
| | - David Nutt
- Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, UK
| | - David Erritzoe
- Centre for Psychedelic Research, Division of Psychiatry, Department of Brain Science, Imperial College of London, UK
| |
Collapse
|
2
|
Chen C, Masotti M, Shepard N, Promes V, Tombesi G, Arango D, Manzoni C, Greggio E, Hilfiker S, Kozorovitskiy Y, Parisiadou L. LRRK2 mediates haloperidol-induced changes in indirect pathway striatal projection neurons. Mol Psychiatry 2025:10.1038/s41380-025-03030-z. [PMID: 40269187 DOI: 10.1038/s41380-025-03030-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025]
Abstract
Haloperidol is used to manage psychotic symptoms in several neurological disorders through mechanisms that involve antagonism of dopamine D2 receptors that are highly expressed in the striatum. Significant side effects of haloperidol, known as extrapyramidal symptoms, lead to motor deficits similar to those seen in Parkinson's disease and present a major challenge in clinical settings. The underlying molecular mechanisms responsible for these side effects remain poorly understood. Parkinson's disease-associated leucine-rich repeat kinase 2 (LRRK2) has an essential role in striatal physiology and a known link to dopamine D2 receptor signaling. Here, we systematically explore convergent signaling of haloperidol and LRRK2 through pharmacological or genetic inhibition of LRRK2 kinase, as well as knock-in mouse models expressing pathogenic mutant LRRK2 with increased kinase activity. Behavioral assays show that LRRK2 kinase inhibition ameliorates haloperidol-induced motor changes in mice. A combination of electrophysiological and anatomical approaches reveals that LRRK2 kinase inhibition interferes with haloperidol-induced changes, specifically in striatal neurons of the indirect pathway. Proteomic studies and targeted intracellular pathway analyses demonstrate that haloperidol induces a similar pattern of intracellular signaling as increased LRRK2 kinase activity. Our study suggests that LRRK2 kinase plays a key role in striatal dopamine D2 receptor signaling underlying the undesirable motor side effects of haloperidol. This work opens up new therapeutic avenues for dopamine-related disorders, such as psychosis, also furthering our understanding of Parkinson's disease pathophysiology.
Collapse
Affiliation(s)
- Chuyu Chen
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Meghan Masotti
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Nathaniel Shepard
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Vanessa Promes
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Giulia Tombesi
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Daniel Arango
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | | | - Elisa Greggio
- Department of Biology, University of Padova, Padova, Italy
| | - Sabine Hilfiker
- Department of Anesthesiology, Rutgers, New Jersey Medical School, Newark, NJ, USA
| | | | - Loukia Parisiadou
- Department of Pharmacology, Northwestern University, Chicago, IL, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
3
|
de la Salle S, Phillips JL, Blier P, Knott V. Acute subanesthetic ketamine-induced effects on the mismatch negativity and their relationship to early and sustained treatment response in major depressive disorder. J Psychopharmacol 2025:2698811251319456. [PMID: 40012166 DOI: 10.1177/02698811251319456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
BACKGROUND A sub-anesthetic dose of ketamine, an N-methyl-D-aspartate receptor (NMDAR) antagonist, produces robust antidepressant effects in treatment-resistant major depressive disorder (MDD). The mismatch negativity (MMN) is reliant on glutamatergic neurotransmission and reduced by NMDAR antagonists. The MMN may characterise the neural mechanisms underlying ketamine's effects. AIMS This study examined the acute effects of ketamine and midazolam on the MMN and its relationship to early and sustained decreases in depressive symptoms. METHODS Treatment-resistant MDD patients (N = 24), enrolled in a multi-phase clinical ketamine trial, received two intravenous infusions within an initial double-blind crossover phase: ketamine (0.5 mg/kg) and midazolam (30 μg/kg). Three recordings were carried out per session (pre-, immediately post- and 2 h post-infusion). Peak MMN amplitude (μV), latency (ms), theta event-related oscillations (EROs), theta phase locking factor (PLF) and source-localised MMN generator activity were assessed. Relationships between changes in MMN indices and early (Phase 1: double-blind, cross-over phase) and sustained (Phases 2, 3: open-label repeated and maintenance phases, respectively) changes in depressive symptoms (Montgomery-Åsberg Depression Rating Scale score) were examined. RESULTS Ketamine reduced frontal MMN amplitudes, theta ERO immediately post- and 2 h post-infusion and source-localised peak MMN frontal generator activity. Select baseline and ketamine-induced MMN decreases correlated and predicted greater early (left frontal MMN decreases in amplitude and theta ERO, baseline left PLF) and sustained (baseline left PLF, right inferior temporal activity) symptom reductions. CONCLUSIONS Acute NMDARs blockade reduced frontal MMN, with larger MMN reductions predicting greater symptom improvement. The MMN may serve as a non-invasive biomarker predicting antidepressant response to glutamatergic agents.
Collapse
Affiliation(s)
- Sara de la Salle
- University of Ottawa Institute of Mental Health Research at the Royal, Ottawa, ON, Canada
- School of Psychology, University of Ottawa, Ottawa, ON, Canada
| | - Jennifer L Phillips
- University of Ottawa Institute of Mental Health Research at the Royal, Ottawa, ON, Canada
- Department of Psychiatry, University of Ottawa, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Pierre Blier
- University of Ottawa Institute of Mental Health Research at the Royal, Ottawa, ON, Canada
- Department of Psychiatry, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Verner Knott
- University of Ottawa Institute of Mental Health Research at the Royal, Ottawa, ON, Canada
- School of Psychology, University of Ottawa, Ottawa, ON, Canada
- Department of Psychiatry, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
4
|
Du Z, Zhu XM, Lv P, Pan Y, Hou XK, Li A, Zhao D, Xing JX, Yao J. Ketamine Alters Specific Gene Expression Profiles by Transcriptome-Wide Responses in a Ketamine-Induced Schizophrenia-Like Mouse Model. Mol Neurobiol 2025:10.1007/s12035-025-04789-6. [PMID: 39992587 DOI: 10.1007/s12035-025-04789-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 02/16/2025] [Indexed: 02/26/2025]
Abstract
Psychotic disorder is a significant consequence of ketamine abuse. However, the molecular mechanisms and biomarkers for this psychotic disorder and associated long-term cognitive impairment remain unclear. To investigate the behavioral changes and comprehensive gene expression alterations in mice following ketamine administration, we employed behavioral testing and RNA sequencing (RNA-seq). We further examined the role of dopamine D1 receptor (Drd1) activity in mediating ketamine-induced psychotic-like behavior and its impact on the transcriptome in these mice. Our findings indicated that blocking Drd1 activity with an antagonist mitigated ketamine-induced schizophrenia-like behaviors, while activating Drd1 with an agonist partially replicated these symptoms. Transcriptome analysis of the mouse hippocampus using RNA-seq revealed an enrichment of differentially expressed genes implicated in the GTPase activation pathway. Specifically, both Rgs4 and Gnai3 were involved in ketamine-induced psychiatric effects. Furthermore, we observed that the mRNA expression of Gnai3 was decreased in peripheral blood and the serum levels of eotaxin-2 were elevated two weeks after ketamine administration. These changes suggest that Gnai3 and eotaxin-2 may serve as potential biomarkers for ketamine abuse. These results demonstrate the crucial role of Drd1 activity in a mouse model of ketamine-induced schizophrenia-like disorder. The altered expression of Gnai3 in peripheral blood and the elevated levels of cytokine eotaxin-2 in serum indicate their potential as peripheral blood biomarkers for ketamine abuse in mice.
Collapse
Affiliation(s)
- Zhe Du
- School of Forensic Medicine, Shenbei New District, China Medical University, No.77, Puhe Road, Shenyang, 110122, P.R. China
- Key Laboratory of Forensic Bio-Evidence Sciences, Liaoning Province, Shenyang, P.R. China
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China
- Langfang Health Vocational College, Langfang, P.R. China
| | - Xiu-Mei Zhu
- School of Forensic Medicine, Shenbei New District, China Medical University, No.77, Puhe Road, Shenyang, 110122, P.R. China
- Key Laboratory of Forensic Bio-Evidence Sciences, Liaoning Province, Shenyang, P.R. China
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China
| | - Peng Lv
- School of Forensic Medicine, Shenbei New District, China Medical University, No.77, Puhe Road, Shenyang, 110122, P.R. China
- Key Laboratory of Forensic Bio-Evidence Sciences, Liaoning Province, Shenyang, P.R. China
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China
| | - Ying Pan
- School of Forensic Medicine, Shenbei New District, China Medical University, No.77, Puhe Road, Shenyang, 110122, P.R. China
- Key Laboratory of Forensic Bio-Evidence Sciences, Liaoning Province, Shenyang, P.R. China
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China
| | - Xi-Kai Hou
- School of Forensic Medicine, Shenbei New District, China Medical University, No.77, Puhe Road, Shenyang, 110122, P.R. China
- Key Laboratory of Forensic Bio-Evidence Sciences, Liaoning Province, Shenyang, P.R. China
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China
| | - Ang Li
- School of Forensic Medicine, Shenbei New District, China Medical University, No.77, Puhe Road, Shenyang, 110122, P.R. China
- Key Laboratory of Forensic Bio-Evidence Sciences, Liaoning Province, Shenyang, P.R. China
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China
| | - Dong Zhao
- Key Laboratory of Evidence Science, China University of Political Science and Law, Ministry of Education, Beijing, P.R. China.
| | - Jia-Xin Xing
- School of Forensic Medicine, Shenbei New District, China Medical University, No.77, Puhe Road, Shenyang, 110122, P.R. China.
- Key Laboratory of Forensic Bio-Evidence Sciences, Liaoning Province, Shenyang, P.R. China.
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China.
| | - Jun Yao
- School of Forensic Medicine, Shenbei New District, China Medical University, No.77, Puhe Road, Shenyang, 110122, P.R. China.
- Key Laboratory of Forensic Bio-Evidence Sciences, Liaoning Province, Shenyang, P.R. China.
- China Medical University Center of Forensic Investigation, Shenyang, P.R. China.
| |
Collapse
|
5
|
Duque M, Chen AB, Hsu E, Narayan S, Rymbek A, Begum S, Saher G, Cohen AE, Olson DE, Li Y, Prober DA, Bergles DE, Fishman MC, Engert F, Ahrens MB. Ketamine induces plasticity in a norepinephrine-astroglial circuit to promote behavioral perseverance. Neuron 2025; 113:426-443.e5. [PMID: 39694033 PMCID: PMC11889991 DOI: 10.1016/j.neuron.2024.11.011] [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: 03/16/2024] [Revised: 08/08/2024] [Accepted: 11/20/2024] [Indexed: 12/20/2024]
Abstract
Transient exposure to ketamine can trigger lasting changes in behavior and mood. We found that brief ketamine exposure causes long-term suppression of futility-induced passivity in larval zebrafish, reversing the "giving-up" response that normally occurs when swimming fails to cause forward movement. Whole-brain imaging revealed that ketamine hyperactivates the norepinephrine-astroglia circuit responsible for passivity. After ketamine washout, this circuit exhibits hyposensitivity to futility, leading to long-term increased perseverance. Pharmacological, chemogenetic, and optogenetic manipulations show that norepinephrine and astrocytes are necessary and sufficient for ketamine's long-term perseverance-enhancing aftereffects. In vivo calcium imaging revealed that astrocytes in adult mouse cortex are similarly activated during futility in the tail suspension test and that acute ketamine exposure also induces astrocyte hyperactivation. The cross-species conservation of ketamine's modulation of noradrenergic-astroglial circuits and evidence that plasticity in this pathway can alter the behavioral response to futility hold promise for identifying new strategies to treat affective disorders.
Collapse
Affiliation(s)
- Marc Duque
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Graduate Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA; Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA.
| | - Alex B Chen
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Graduate Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA; Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA.
| | - Eric Hsu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sujatha Narayan
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Altyn Rymbek
- Tianqiao and Chrissy Chen Institute for Neuroscience, Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Shahinoor Begum
- Department of Physics, Harvard University, Cambridge, MA 02138, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Gesine Saher
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen 37075, Germany
| | - Adam E Cohen
- Department of Physics, Harvard University, Cambridge, MA 02138, USA; Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - David E Olson
- Department of Chemistry, University of California, Davis, Davis, CA 95616, USA; Department of Biochemistry & Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA; Center for Neuroscience, University of California, Davis, Davis, CA 95618, USA; Institute for Psychedelics and Neurotherapeutics, University of California, Davis, Davis, CA 95616, USA
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing 100871, China; PKU-IDG/McGovern Institute for Brain Research, Beijing 100871, China
| | - David A Prober
- Tianqiao and Chrissy Chen Institute for Neuroscience, Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Dwight E Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Mark C Fishman
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Florian Engert
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Misha B Ahrens
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA.
| |
Collapse
|
6
|
Liao C, Dua AN, Wojtasiewicz C, Liston C, Kwan AC. Structural neural plasticity evoked by rapid-acting antidepressant interventions. Nat Rev Neurosci 2025; 26:101-114. [PMID: 39558048 PMCID: PMC11892022 DOI: 10.1038/s41583-024-00876-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2024] [Indexed: 11/20/2024]
Abstract
A feature in the pathophysiology of major depressive disorder (MDD), a mood disorder, is the impairment of excitatory synapses in the prefrontal cortex. Intriguingly, different types of treatment with fairly rapid antidepressant effects (within days or a few weeks), such as ketamine, electroconvulsive therapy and non-invasive neurostimulation, seem to converge on enhancement of neural plasticity. However, the forms and mechanisms of plasticity that link antidepressant interventions to the restoration of excitatory synaptic function are still unknown. In this Review, we highlight preclinical research from the past 15 years showing that ketamine and psychedelic drugs can trigger the growth of dendritic spines in cortical pyramidal neurons. We compare the longitudinal effects of various psychoactive drugs on neuronal rewiring, and we highlight rapid onset and sustained time course as notable characteristics for putative rapid-acting antidepressant drugs. Furthermore, we consider gaps in the current understanding of drug-evoked in vivo structural plasticity. We also discuss the prospects of using synaptic remodelling to understand other antidepressant interventions, such as repetitive transcranial magnetic stimulation. Finally, we conclude that structural neural plasticity can provide unique insights into the neurobiological actions of psychoactive drugs and antidepressant interventions.
Collapse
Affiliation(s)
- Clara Liao
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, CT, USA
| | - Alisha N Dua
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
| | | | - Conor Liston
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Alex C Kwan
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
- Department of Psychiatry, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
7
|
Zahid Z, Sultan ZW, Krause BM, Wenthur CJ, Pearce RA, Banks MI. Divergent Effects of Ketamine and the Serotoninergic Psychedelic 2,5-Dimethoxy-4-Iodoamphetamine on Hippocampal Plasticity and Metaplasticity. PSYCHEDELIC MEDICINE (NEW ROCHELLE, N.Y.) 2024; 2:166-177. [PMID: 39669671 PMCID: PMC11633440 DOI: 10.1089/psymed.2023.0061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2024]
Abstract
Introduction Serotonergic psychedelics and ketamine produce rapid and long-lasting symptomatic relief in multiple psychiatric disorders. Evidence suggests that despite having distinct molecular targets, both drugs may exert therapeutic benefit via their pro-neuroplastic effects. Following treatment with ketamine or serotonergic psychedelics, patients are reported to be more open to behavioral change, which is leveraged for psychotherapy-assisted reframing of narratives of the self. This period of enhanced behavioral change is postulated to be supported by a post-treatment window of enhanced neural plasticity, but evidence for such 'metaplastic' effects is limited. In this study, we tested for neural plasticity and metaplasticity in murine hippocampus. Methods Brain slices were obtained from C57BL/6J mice 24 hours after treatment (intraperitoneal injection) with saline, ketamine, or the serotonergic psychedelic 2,5-Dimethoxy-4-iodoamphetamine (DOI). Extracellular fiber volleys (FVs) and field excitatory postsynaptic potentials (fEPSPs) were recorded in stratum radiatum of CA1 in response to stimulation of Schaffer collateral fibers before and after induction of short-term and long-term potentiation (STP, LTP). Results Before LTP induction, responses differed across treatment groups (F1,2 = 5.407, p = 0.00665), with fEPSPs enhanced in slices from DOI-treated animals (p = 0.0182), but not ketamine-treated animals (p = 0.9786), compared to saline. There were no treatment effects on LT (F1,2 = 0.6, p = 0.516), but there were on STP (F1,2 =, p = 0.0167), with enhanced STP in DOI-treated (p = 0.0352) but not ketamine-treated (p = 0.9999) animals compared to saline. A presynaptic component to the mechanism for the DOI effects was suggested by (1) significantly enhanced FV amplitudes (F1,2 = 3.17, p = 0.049) in DOI-treated (p = 0.0457) but not ketamine-treated animals compared to saline (p = 0.8677); and (2) enhanced paired pulse ratios (F1,2 = 3.581, p = 0.0339) in slices from DOI-treated (p= 0.0257) but not ketamine-treated animals (p = 0.4845) compared to saline. Conclusions DOI, but not ketamine, induced significant neuroplastic and metaplastic effects at hippocampal CA1 synapses 24 hours after treatment, likely in part via a presynaptic mechanism.
Collapse
Affiliation(s)
- Zarmeen Zahid
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706
- School of Pharmacy, University of Wisconsin, Madison, WI, 53705
- Neuroscience Training Program, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706
| | - Ziyad W. Sultan
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706
| | - Bryan M. Krause
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706
| | - Cody J. Wenthur
- School of Pharmacy, University of Wisconsin, Madison, WI, 53705
- Transdisciplinary Center for Research in Psychoactive Substances, University of Wisconsin, Madison, WI, 53705
- Neuroscience Training Program, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706
| | - Robert A. Pearce
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706
| | - Matthew I. Banks
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706
- Transdisciplinary Center for Research in Psychoactive Substances, University of Wisconsin, Madison, WI, 53705
- Neuroscience Training Program, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53706
| |
Collapse
|
8
|
Krüssel S, Deb I, Son S, Ewall G, Chang M, Lee HK, Heo WD, Kwon HB. H-Ras induces exuberant de novo dendritic protrusion growth in mature neurons regardless of cell type. iScience 2024; 27:110535. [PMID: 39220408 PMCID: PMC11365382 DOI: 10.1016/j.isci.2024.110535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 05/03/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024] Open
Abstract
Dendritic protrusions, mainly spines and filopodia, correlate with excitatory synapses in pyramidal neurons (PyNs), but this relationship may not apply universally. We found that ectopic H-Ras expression increased protrusions across various cortical cell types, including layer 2/3 PyNs, parvalbumin (PV)-, and vasoactive intestinal peptide (VIP)-positive interneurons (INs) in the primary motor cortex. The probability of detecting protrusions correlated with local H-Ras activity, indicating its role in protrusion formation. H-Ras overexpression led to high turnover rates by adding protrusions. Two-photon photolysis of glutamate induced de novo spine formation in mature H-Ras expressing neurons, suggesting H-Ras's effect is not limited to early development. In PyNs and PV-INs, but not VIP-INs, spine neck lengths shifted to filopodia-like phenotypes. H-Ras primarily induced filopodia in PyNs and spines in PV- and VIP-INs. Increased protrusions in H-Ras-transfected PyNs lacked key excitatory synaptic proteins and did not affect miniature excitatory postsynaptic currents (mEPSCs), suggesting multifaceted roles beyond excitatory synapses.
Collapse
Affiliation(s)
- Sarah Krüssel
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ishana Deb
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Seungkyu Son
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Gabrielle Ewall
- Solomon H. Snyder Department of Neuroscience, Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Minhyeok Chang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hey-Kyoung Lee
- Solomon H. Snyder Department of Neuroscience, Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Won Do Heo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Hyung-Bae Kwon
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
9
|
Wang K, Tan X, Ding KM, Feng XZ, Zhao YY, Zhu WL, Li GH, Li SX. Dynamic regulation of phosphorylation of NMDA receptor GluN2B subunit tyrosine residues mediates ketamine rapid antidepressant effects. Pharmacol Res 2024; 205:107236. [PMID: 38797358 DOI: 10.1016/j.phrs.2024.107236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
The rapid antidepressant effects of ketamine depend on the N-methyl-D-aspartate (NMDA) receptor containing 2B subunit (NR2B), whose function is influenced by its phosphorylated regulation and distribution within and outside synapses. It remains unclear if ketamine's rapid onset of antidepressant effects relies on the dynamic phosphorylated regulation of NR2B within and outside synapses. Here, we show that ketamine rapidlyalleviated depression-like behaviors and normalized abnormal expression of pTyr1472NR2B and striatal-enriched protein tyrosine phosphatase (STEP) 61 within and outside synapses in the medial prefrontal cortex (mPFC) induced by chronic unpredictable stress (CUS) and conditional knockdown of STEP 61, a key phosphatase of NR2B, within 1 hour after administration Together, our results delineate the rapid initiation of ketamine's antidepressant effects results from the restoration of NR2B phosphorylation homeostasis within and outside synapses. The dynamic regulation of phosphorylation of NR2B provides a new perspective for developing new antidepressant strategies.
Collapse
Affiliation(s)
- Ke Wang
- National Institute on Drug Dependence and Beijing Key laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Department of Pharmacology, Peking University Health Science Center, Beijing 100191, China
| | - Xuan Tan
- National Institute on Drug Dependence and Beijing Key laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Department of Neurobiology, Peking University Health Science Center, Beijing 100191, China
| | - Kai-Mo Ding
- National Institute on Drug Dependence and Beijing Key laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Zhenjiang Mental Health Center, Jiangsu 212000, China
| | - Xue-Zhu Feng
- National Institute on Drug Dependence and Beijing Key laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Department of Neurobiology, Peking University Health Science Center, Beijing 100191, China
| | - Yu-Yu Zhao
- National Institute on Drug Dependence and Beijing Key laboratory of Drug Dependence Research, Peking University, Beijing 100191, China; Department of Neurobiology, Peking University Health Science Center, Beijing 100191, China
| | - Wei-Li Zhu
- National Institute on Drug Dependence and Beijing Key laboratory of Drug Dependence Research, Peking University, Beijing 100191, China
| | - Guo-Hai Li
- Zhenjiang Mental Health Center, Jiangsu 212000, China
| | - Su-Xia Li
- National Institute on Drug Dependence and Beijing Key laboratory of Drug Dependence Research, Peking University, Beijing 100191, China.
| |
Collapse
|
10
|
Bærentzen SL, Thomsen JB, Thomsen MB, Jakobsen S, Simonsen MT, Wegener G, Brooks DJ, Landau AM. Subanesthetic S-ketamine does not acutely alter striatal dopamine transporter binding in healthy Sprague Dawley female rats. Synapse 2024; 78:e22294. [PMID: 38813759 DOI: 10.1002/syn.22294] [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: 01/17/2024] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/31/2024]
Abstract
Major depressive disorder is one of the most prevalent mental health disorders, posing a global socioeconomic burden. Conventional antidepressant treatments have a slow onset of action, and 30% of patients show no clinically significant treatment response. The recently approved fast-acting antidepressant S-ketamine, an N-methyl-D-aspartate receptor antagonist, provides a new approach for treatment-resistant patients. However, knowledge of S-ketamine's mechanism of action is still being established. Depressed human subjects have lower striatal dopamine transporter (DAT) availability compared to healthy controls. Rodent studies report increased striatal dopamine concentration in response to acute ketamine administration. In vivo [18F]FE-PE2I ([18F]-(E)-N-(3-iodoprop-2-enyl)-2β-carbofluoroethoxy-3β-(4'-methyl-phenyl) nortropane) positron emission tomography (PET) imaging of the DAT has not previously been applied to assess the effect of acute subanesthetic S-ketamine administration on DAT availability. We applied translational in vivo [18F]FE-PE2I PET imaging of the DAT in healthy female rats to evaluate whether an acute subanesthetic intraperitoneal dose of 15 mg/kg S-ketamine alters DAT availability. We also performed [3H]GBR-12935 autoradiography on postmortem brain sections. We found no effect of acute S-ketamine administration on striatal DAT binding using [18F]FE-PE2I PET or [3H]GBR-12935 autoradiography. This negative result does not support the hypothesis that DAT changes are associated with S-ketamine's rapid antidepressant effects, but additional studies are warranted.
Collapse
Affiliation(s)
- Simone Larsen Bærentzen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jakob Borup Thomsen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Majken Borup Thomsen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Steen Jakobsen
- Department of Nuclear Medicine and PET Centre, Aarhus University and Hospital, Aarhus, Denmark
| | | | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - David J Brooks
- Department of Nuclear Medicine and PET Centre, Aarhus University and Hospital, Aarhus, Denmark
- Institute of Translational and Clinical Research, University of Newcastle upon Tyne, Newcastle upon Tyne, UK
| | - Anne M Landau
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
11
|
Lan J, Chen Z, Cheng H. Dopamine release after acute sleep deprivation: culprit of affective state transitions. MedComm (Beijing) 2024; 5:e630. [PMID: 38952576 PMCID: PMC11215283 DOI: 10.1002/mco2.630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/17/2024] [Accepted: 06/04/2024] [Indexed: 07/03/2024] Open
Affiliation(s)
- Ju Lan
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), School of Pharmaceutical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), School of Pharmaceutical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| | - Heming Cheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), School of Pharmaceutical SciencesZhejiang Chinese Medical UniversityHangzhouChina
| |
Collapse
|
12
|
Torrado Pacheco A, Moghaddam B. Licit use of illicit drugs for treating depression: the pill and the process. J Clin Invest 2024; 134:e180217. [PMID: 40047885 PMCID: PMC11178541 DOI: 10.1172/jci180217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2025] Open
Abstract
Psilocybin, MDMA, and ketamine have emerged as potentially effective treatments for rapid amelioration of the symptoms of mood and related psychiatric disorders. All clinical data collected so far with regard to psilocybin or MDMA, which have reported positive outcomes for treating depression, anxiety, posttraumatic stress disorder, and drug or alcohol use disorders, have involved clinician-assisted intervention. While the case for ketamine is assumed to be different, the first report of the successful use of ketamine in psychiatry for treating depression was in combination with psychotherapy, and an emerging literature suggests that the subjective state of individual experiences with ketamine predicts clinical outcome. This Review will focus on (a) a brief review of the literature, showing that the context or the process of drug administration has been an integrative component of published work; (b) the importance of clinical trials to compare the efficacy of the drug ("pill") as a stand-alone treatment versus drug in combination with clinician-assisted psychological support ("process"); and (c) suggestions for future approaches in animal models that take into account the role of systems and behavioral neuroscience in explaining a potential role for context, experience, and expectancy in drug effect.
Collapse
Affiliation(s)
| | - Bita Moghaddam
- Department of Behavioral Neuroscience, and
- Department of Psychiatry, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
13
|
Chen C, Masotti M, Shepard N, Promes V, Tombesi G, Arango D, Manzoni C, Greggio E, Hilfiker S, Kozorovitskiy Y, Parisiadou L. LRRK2 mediates haloperidol-induced changes in indirect pathway striatal projection neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597594. [PMID: 38895420 PMCID: PMC11185612 DOI: 10.1101/2024.06.06.597594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Haloperidol is used to manage psychotic symptoms in several neurological disorders through mechanisms that involve antagonism of dopamine D2 receptors that are highly expressed in the striatum. Significant side effects of haloperidol, known as extrapyramidal symptoms, lead to motor deficits similar to those seen in Parkinson's disease and present a major challenge in clinical settings. The underlying molecular mechanisms responsible for these side effects remain poorly understood. Parkinson's disease-associated LRRK2 kinase has an important role in striatal physiology and a known link to dopamine D2 receptor signaling. Here, we systematically explore convergent signaling of haloperidol and LRRK2 through pharmacological or genetic inhibition of LRRK2 kinase, as well as knock-in mouse models expressing pathogenic mutant LRRK2 with increased kinase activity. Behavioral assays show that LRRK2 kinase inhibition ameliorates haloperidol-induced motor changes in mice. A combination of electrophysiological and anatomical approaches reveals that LRRK2 kinase inhibition interferes with haloperidol-induced changes, specifically in striatal neurons of the indirect pathway. Proteomic studies and targeted intracellular pathway analyses demonstrate that haloperidol induces a similar pattern of intracellular signaling as increased LRRK2 kinase activity. Our study suggests that LRRK2 kinase plays a key role in striatal dopamine D2 receptor signaling underlying the undesirable motor side effects of haloperidol. This work opens up new therapeutic avenues for dopamine-related disorders, such as psychosis, also furthering our understanding of Parkinson's disease pathophysiology.
Collapse
Affiliation(s)
- Chuyu Chen
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Meghan Masotti
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Nathaniel Shepard
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Vanessa Promes
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Giulia Tombesi
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Daniel Arango
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | | | | | - Sabine Hilfiker
- Department of Anesthesiology, Rutgers, New Jersey Medical School, NJ, USA
| | | | - Loukia Parisiadou
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| |
Collapse
|
14
|
Bhatti DL, Jin J, Cheng J, McCabe K, Lee KW, Berdasco C, Jeong YY, Sinha SC, Kim Y. Ahnak in the prefrontal cortex mediates behavioral correlates of stress resilience and rapid antidepressant action in mice. Front Mol Neurosci 2024; 17:1350716. [PMID: 38828281 PMCID: PMC11140847 DOI: 10.3389/fnmol.2024.1350716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/12/2024] [Indexed: 06/05/2024] Open
Abstract
The prefrontal cortex (PFC) is a key neural node mediating behavioral responses to stress and the actions of ketamine, a fast-acting antidepressant. The molecular mechanisms underlying these processes, however, are not fully understood. Our recent study revealed a pivotal role of hippocampal Ahnak as a regulator of cellular and behavioral adaptations to chronic stress. However, despite its significant expression in the PFC, the contribution of cortical Ahnak to behavioral responses to stress and antidepressants remains unknown. Here, using a mouse model for chronic social stress, we find that Ahnak expression in the PFC is significantly increased in stress-resilient mice and positively correlated with social interaction after stress exposure. Conditional deletion of Ahnak in the PFC or forebrain glutamatergic neurons facilitates stress susceptibility, suggesting that Ahnak is required for behavioral resilience. Further supporting this notion, Ahnak expression in the PFC is increased after the administration of ketamine or its metabolite (2R, 6R)-hydroxynorketamine (HNK). Moreover, Ahnak deletion in forebrain glutamatergic neurons blocks the restorative behavioral effects of ketamine or HNK in stress-susceptible mice. This forebrain excitatory neuron-specific Ahnak deletion reduces the frequency of mini excitatory postsynaptic currents in layer II/III pyramidal neurons, suggesting that Ahnak may induce its behavioral effects via modulation of glutamatergic transmission in the PFC. Altogether, these data suggest that Ahnak in glutamatergic PFC neurons may be critical for behavioral resilience and antidepressant actions of ketamine or HNK in chronic social stress-exposed mice.
Collapse
Affiliation(s)
- Dionnet L. Bhatti
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
- Program in Neuroscience, Harvard Medical School, Boston, MA, United States
| | - Junghee Jin
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Jia Cheng
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Kathryn McCabe
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Ko-Woon Lee
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
| | - Clara Berdasco
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States
| | - Yu Young Jeong
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States
| | - Subhash C. Sinha
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
- Weill Cornell Medicine Helen & Robert Appel Alzheimer’s Disease Research Institute, New York, NY, United States
| | - Yong Kim
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, United States
- Department of Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States
- Brain Health Institute, Rutgers University, Piscataway, NJ, United States
| |
Collapse
|
15
|
Yi L, Ma H, Yang X, Zheng Q, Zhong J, Ye S, Li X, Chen D, Li H, Li C. Cotransplantation of NSCs and ethyl stearate promotes synaptic plasticity in PD rats by Drd1/ERK/AP-1 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117292. [PMID: 37806537 DOI: 10.1016/j.jep.2023.117292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 10/04/2023] [Accepted: 10/06/2023] [Indexed: 10/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine views kidney shortage as a significant contributor to the aetiology of Parkinson's disease (PD), a neurodegenerative condition that is closely linked to aging. In clinical, patients with Parkinson's disease are often treated with Testudinis Carapax et Plastrum (Plastrum Testudinis, PT), a traditional Chinese medication that tonifies the kidney. Previous research has demonstrated that ethyl stearate (PubChem CID: 8122), an active component of Plastrum Testudinis Extracted with ethyl acetate (PTE), may encourage neural stem cells (NSCs) development into dopaminergic (DAergic) neurons. However, the effectiveness and mechanism of cotransplantation of ethyl stearate and NSCs in treating PD model rats still require further investigation. AIM OF THE STUDY PD is a neurodegenerative condition marked by the loss and degradation of dopaminergic neurons in the substantia nigra of the midbrain. Synaptic damage is also a critical pathology in PD. Because of their self-renewal, minimal immunogenicity, and capacity to differentiate into dopaminergic (DAergic) neurons, NSCs are a prospective treatment option for Parkinson's disease cell transplantation therapy. However, encouraging transplanted NSCs to differentiate into dopaminergic neurons and enhancing synaptic plasticity in vivo remains a significant challenge in improving the efficacy of NSCs transplantation for PD. This investigation seeks to examine the efficacy of cotransplantation of NSCs and ethyl stearate in PD model rats and its mechanism related to synaptic plasticity. MATERIALS AND METHODS On 6-hydroxydopamine-induced PD model rats, we performed NSCs transplantation therapy and cotransplantation therapy involving ethyl stearate and NSCs. Rotating behavior induced by apomorphine (APO) and pole climbing tests were used to evaluate behavioral changes. Using a variety of methods, including Western blotting (WB), immunofluorescence analysis, enzyme-linked immunosorbent assay, and quantitative real-time polymerase chain reaction (qRT-PCR), we examined the function and potential molecular mechanisms of ethyl stearate in combined NSCs transplantation therapy. RESULTS In the rat PD model, cotransplantation of ethyl stearate with NSCs dramatically reduced motor dysfunction, restored TH protein levels, and boosted dopamine levels in the striatum, according to our findings. Furthermore, the expression levels of SYN1 and PSD95, markers of synaptic plasticity, and BDNF, closely related to synaptic plasticity, were significantly increased. Cotransplantation with ethyl stearate and NSCs also increased the expression levels of Dopamine Receptor D1 (Drd1), an important receptor in the dopamine neural circuit, accompanied by an increase in MMP9 levels, ERK1/2 phosphorylation levels, and c-fos protein levels. CONCLUSIONS According to the results of our investigation, cotransplantation of ethyl stearate and NSCs significantly improves the condition of PD model rats. We found that cotransplantation of ethyl stearate and NSCs may promote the expression of MMP9 by regulating the Drd1-ERK-AP-1 pathway, thus improving synaptic plasticity after NSCs transplantation. These findings provide new experimental support for the treatment of PD with the kidney tonifying Chinese medicine Plastrum Testudinis and suggest a potential therapeutic strategy for PD based on cotransplantation therapy.
Collapse
Affiliation(s)
- Lan Yi
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China; Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China; Guangzhou Huaxia Vocational College, Guangzhou, Guangdong Province, 510935, PR China
| | - Haisheng Ma
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China; Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China
| | - Xiaoxiao Yang
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China; Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China
| | - Qi Zheng
- School of Information Science and Technology, Guangdong University of Foreign Studies, Guangzhou, Guangdong Province, 510006, PR China
| | - Jun Zhong
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China; Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China
| | - Sen Ye
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China; Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China
| | - Xican Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China
| | - Dongfeng Chen
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China
| | - Hui Li
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China.
| | - Caixia Li
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China; Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, 510006, PR China.
| |
Collapse
|
16
|
Evans JW, Graves MC, Nugent AC, Zarate CA. Hippocampal volume changes after (R,S)-ketamine administration in patients with major depressive disorder and healthy volunteers. Sci Rep 2024; 14:4538. [PMID: 38402253 PMCID: PMC10894199 DOI: 10.1038/s41598-024-54370-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 02/12/2024] [Indexed: 02/26/2024] Open
Abstract
The hippocampus and amygdala have been implicated in the pathophysiology and treatment of major depressive disorder (MDD). Preclinical models suggest that stress-related changes in these regions can be reversed by antidepressants, including ketamine. Clinical studies have identified reduced volumes in MDD that are thought to be potentiated by early life stress and worsened by repeated depressive episodes. This study used 3T and 7T structural magnetic resonance imaging data to examine longitudinal changes in hippocampal and amygdalar subfield volumes associated with ketamine treatment. Data were drawn from a previous double-blind, placebo-controlled, crossover trial of healthy volunteers (HVs) unmedicated individuals with treatment-resistant depression (TRD) (3T: 18 HV, 26 TRD, 7T: 17 HV, 30 TRD) who were scanned at baseline and twice following either a 40 min IV ketamine (0.5 mg/kg) or saline infusion (acute: 1-2 days, interim: 9-10 days post infusion). No baseline differences were noted between the two groups. At 10 days post-infusion, a slight increase was observed between ketamine and placebo scans in whole left amygdalar volume in individuals with TRD. No other differences were found between individuals with TRD and HVs at either field strength. These findings shed light on the timing of ketamine's effects on cortical structures.
Collapse
Affiliation(s)
- Jennifer W Evans
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr., Bldg 10, Rm 7-3335, Bethesda, MD, 20814, USA.
| | - Morgan C Graves
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr., Bldg 10, Rm 7-3335, Bethesda, MD, 20814, USA
| | - Allison C Nugent
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr., Bldg 10, Rm 7-3335, Bethesda, MD, 20814, USA
- MEG Core, NIMH, Bethesda, MD, USA
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr., Bldg 10, Rm 7-3335, Bethesda, MD, 20814, USA
| |
Collapse
|
17
|
Ren L. The mechanistic basis for the rapid antidepressant-like effects of ketamine: From neural circuits to molecular pathways. Prog Neuropsychopharmacol Biol Psychiatry 2024; 129:110910. [PMID: 38061484 DOI: 10.1016/j.pnpbp.2023.110910] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
Conventional antidepressants that target monoaminergic receptors require several weeks to be efficacious. This lag represents a significant problem in the currently available treatments for serious depression. Ketamine, acting as an N-methyl-d-aspartate receptor antagonist, was shown to have rapid antidepressant-like effects, marking a significant advancement in the study of mood disorders. However, serious side effects and adverse reactions limit its clinical use. Considering the limitations of ketamine, it is crucial to further define the network targets of ketamine. The rapid action of ketamine an as antidepressant is thought to be mediated by the glutamate system. It is believed that synaptic plasticity is essential for the rapid effects of ketamine as an antidepressant. Other mechanisms include the involvement of the γ-aminobutyric acidergic (GABAergic), 5-HTergic systems, and recent studies have linked astrocytes to ketamine's rapid antidepressant-like effects. The interactions between these systems exert a synergistic rapid antidepressant effect through neural circuits and molecular mechanisms. Here, we discuss the neural circuits and molecular mechanisms underlying the action of ketamine. This work will help explain how molecular and neural targets are responsible for the effects of rapidly acting antidepressants and will aid in the discovery of new therapeutic approaches for major depressive disorder.
Collapse
Affiliation(s)
- Li Ren
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu 611137, China.
| |
Collapse
|
18
|
Sulaman BA, Kudlak T, Eban-Rothschild A. Dopamine's reach: Unlocked by sleep loss. Neuron 2024; 112:4-6. [PMID: 38176390 DOI: 10.1016/j.neuron.2023.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 01/06/2024]
Abstract
In this issue of Neuron, Wu et al.1 employ cutting-edge techniques to provide a mechanistic understanding of how sleep deprivation induces an altered affective state. They reveal a key function for dopaminergic signaling, and the formation of cortical spines, in this process.
Collapse
Affiliation(s)
- Bibi Alika Sulaman
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA; Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Tyler Kudlak
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA
| | | |
Collapse
|
19
|
Wu M, Zhang X, Feng S, Freda SN, Kumari P, Dumrongprechachan V, Kozorovitskiy Y. Dopamine pathways mediating affective state transitions after sleep loss. Neuron 2024; 112:141-154.e8. [PMID: 37922904 PMCID: PMC10841919 DOI: 10.1016/j.neuron.2023.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 07/25/2023] [Accepted: 10/02/2023] [Indexed: 11/07/2023]
Abstract
The pathophysiology of affective disorders-particularly circuit-level mechanisms underlying bidirectional, periodic affective state transitions-remains poorly understood. In patients, disruptions of sleep and circadian rhythm can trigger transitions to manic episodes, whereas depressive states are reversed. Here, we introduce a hybrid automated sleep deprivation platform to induce transitions of affective states in mice. Acute sleep loss causes mixed behavioral states, featuring hyperactivity, elevated social and sexual behaviors, and diminished depressive-like behaviors, where transitions depend on dopamine (DA). Using DA sensor photometry and projection-targeted chemogenetics, we reveal that elevated DA release in specific brain regions mediates distinct behavioral changes in affective state transitions. Acute sleep loss induces DA-dependent enhancement in dendritic spine density and uncaging-evoked dendritic spinogenesis in the medial prefrontal cortex, whereas optically mediated disassembly of enhanced plasticity reverses the antidepressant effects of sleep deprivation on learned helplessness. These findings demonstrate that brain-wide dopaminergic pathways control sleep-loss-induced polymodal affective state transitions.
Collapse
Affiliation(s)
- Mingzheng Wu
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL 60208, USA
| | - Xin Zhang
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Sihan Feng
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Sara N Freda
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Pushpa Kumari
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Vasin Dumrongprechachan
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA
| | - Yevgenia Kozorovitskiy
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
20
|
Riggs LM, Pereira EFR, Thompson SM, Gould TD. cAMP-dependent protein kinase signaling is required for ( 2R,6R)-hydroxynorketamine to potentiate hippocampal glutamatergic transmission. J Neurophysiol 2024; 131:64-74. [PMID: 38050689 PMCID: PMC11286304 DOI: 10.1152/jn.00326.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/10/2023] [Accepted: 11/23/2023] [Indexed: 12/06/2023] Open
Abstract
(2R,6R)-Hydroxynorketamine (HNK) is a ketamine metabolite that shows rapid antidepressant-like effects in preclinical studies and lacks the adverse N-methyl-d-aspartate receptor (NMDAR) inhibition-related properties of ketamine. Investigating how (2R,6R)-HNK exerts its antidepressant actions may be informative in the design of novel pharmacotherapies with improved safety and efficacy. We sought to identify the molecular substrates through which (2R,6R)-HNK induces functional changes at excitatory synapses, a prevailing hypothesis for how rapid antidepressant effects are initiated. We recorded excitatory postsynaptic potentials in hippocampal slices from male Wistar Kyoto rats, which have impaired hippocampal plasticity and are resistant to traditional antidepressants. (2R,6R)-HNK (10 µM) led to a rapid potentiation of electrically evoked excitatory postsynaptic potentials at Schaffer collateral CA1 stratum radiatum synapses. This potentiation was associated with a decrease in paired pulse facilitation, suggesting an increase in the probability of glutamate release. The (2R,6R)-HNK-induced potentiation was blocked by inhibiting either cyclic adenosine monophosphate (cAMP) or its downstream target, cAMP-dependent protein kinase (PKA). As cAMP is a potent regulator of brain-derived neurotrophic factor (BDNF) release, we assessed whether (2R,6R)-HNK exerts this acute potentiation through a rapid increase in cAMP-dependent BDNF-TrkB signaling. We found that the cAMP-PKA-dependent potentiation was not dependent on TrkB activation by BDNF, which functionally delimits the acute synaptic effects of (2R,6R)-HNK from its sustained BDNF-dependent actions in vivo. These results suggest that, by potentiating glutamate release via cAMP-PKA signaling, (2R,6R)-HNK initiates acute adaptations in fast excitatory synaptic transmission that promote structural plasticity leading to maintained antidepressant action.NEW & NOTEWORTHY Ketamine is a rapid-acting antidepressant and its preclinical effects are mimicked by its (2R,6R)-(HNK) metabolite. We found that (2R,6R)-HNK initiates acute adaptations in fast excitatory synaptic transmission by potentiating glutamate release via cAMP-PKA signaling at hippocampal Schaffer collateral synapses. This cAMP-PKA-dependent potentiation was not dependent on TrkB activation by BDNF, which functionally delimits the rapid synaptic effects of (2R,6R)-HNK from its sustained BDNF-dependent actions that are thought to maintain antidepressant action in vivo.
Collapse
Affiliation(s)
- Lace M Riggs
- Program in Neuroscience and Training Program in Integrative Membrane Biology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Edna F R Pereira
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Scott M Thompson
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Todd D Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
- Veterans Affairs Maryland Health Care System, Baltimore, Maryland, United States
| |
Collapse
|
21
|
Huang K, Duan J, Wang R, Ying H, Feng Q, Zhu B, Yang C, Yang L. Landscape of gut microbiota and metabolites and their interaction in comorbid heart failure and depressive symptoms: a random forest analysis study. mSystems 2023; 8:e0051523. [PMID: 37882579 PMCID: PMC10734515 DOI: 10.1128/msystems.00515-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 08/02/2023] [Indexed: 10/27/2023] Open
Abstract
IMPORTANCE There is increasing evidence that alterations in gut microbial composition and function are associated with cardiovascular or psychiatric disease. Therefore, it is meaningful to investigate the taxonomic and functional characterization of the microbiota in HF patients who also have depressive symptoms. In this cross-sectional study, Cloacibacillus and alpha-tocopherol were determined as new diagnostic markers. Furthermore, intestinal microecosystem disorders are closely linked to depressive symptoms in HF patients, providing a new reference viewpoint for understanding the gut-heart/brain axis.
Collapse
Affiliation(s)
- Kai Huang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jiahao Duan
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ruting Wang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Hangfeng Ying
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Qinwen Feng
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Bin Zhu
- Department of Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ling Yang
- Department of Cardiology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
22
|
Bottemanne H, Berkovitch L, Gauld C, Balcerac A, Schmidt L, Mouchabac S, Fossati P. Storm on predictive brain: A neurocomputational account of ketamine antidepressant effect. Neurosci Biobehav Rev 2023; 154:105410. [PMID: 37793581 DOI: 10.1016/j.neubiorev.2023.105410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 08/24/2023] [Accepted: 09/26/2023] [Indexed: 10/06/2023]
Abstract
For the past decade, ketamine, an N-methyl-D-aspartate receptor (NMDAr) antagonist, has been considered a promising treatment for major depressive disorder (MDD). Unlike the delayed effect of monoaminergic treatment, ketamine may produce fast-acting antidepressant effects hours after a single administration at subanesthetic dose. Along with these antidepressant effects, it may also induce transient dissociative (disturbing of the sense of self and reality) symptoms during acute administration which resolve within hours. To understand ketamine's rapid-acting antidepressant effect, several biological hypotheses have been explored, but despite these promising avenues, there is a lack of model to understand the timeframe of antidepressant and dissociative effects of ketamine. In this article, we propose a neurocomputational account of ketamine's antidepressant and dissociative effects based on the Predictive Processing (PP) theory, a framework for cognitive and sensory processing. PP theory suggests that the brain produces top-down predictions to process incoming sensory signals, and generates bottom-up prediction errors (PEs) which are then used to update predictions. This iterative dynamic neural process would relies on N-methyl-D-aspartate (NMDAr) and α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic receptors (AMPAr), two major component of the glutamatergic signaling. Furthermore, it has been suggested that MDD is characterized by over-rigid predictions which cannot be updated by the PEs, leading to miscalibration of hierarchical inference and self-reinforcing negative feedback loops. Based on former empirical studies using behavioral paradigms, neurophysiological recordings, and computational modeling, we suggest that ketamine impairs top-down predictions by blocking NMDA receptors, and enhances presynaptic glutamate release and PEs, producing transient dissociative symptoms and fast-acting antidepressant effect in hours following acute administration. Moreover, we present data showing that ketamine may enhance a delayed neural plasticity pathways through AMPAr potentiation, triggering a prolonged antidepressant effect up to seven days for unique administration. Taken together, the two sides of antidepressant effects with distinct timeframe could constitute the keystone of antidepressant properties of ketamine. These PP disturbances may also participate to a ketamine-induced time window of mental flexibility, which can be used to improve the psychotherapeutic process. Finally, these proposals could be used as a theoretical framework for future research into fast-acting antidepressants, and combination with existing antidepressant and psychotherapy.
Collapse
Affiliation(s)
- Hugo Bottemanne
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225 / UMRS 1127, Sorbonne University / CNRS / INSERM, Paris, France; Sorbonne University, Department of Philosophy, Science Norm Democracy Research Unit, UMR, 8011, Paris, France; Sorbonne University, Department of Psychiatry, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.
| | - Lucie Berkovitch
- Saclay CEA Centre, Neurospin, Gif-Sur-Yvette Cedex, France; Department of Psychiatry, GHU Paris Psychiatrie et Neurosciences, Service Hospitalo-Universitaire, Paris, France
| | - Christophe Gauld
- Department of Child Psychiatry, CHU de Lyon, F-69000 Lyon, France; Institut des Sciences Cognitives Marc Jeannerod, UMR 5229 CNRS & Université Claude Bernard Lyon 1, F-69000 Lyon, France
| | - Alexander Balcerac
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225 / UMRS 1127, Sorbonne University / CNRS / INSERM, Paris, France; Sorbonne University, Department of Neurology, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Liane Schmidt
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225 / UMRS 1127, Sorbonne University / CNRS / INSERM, Paris, France
| | - Stephane Mouchabac
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225 / UMRS 1127, Sorbonne University / CNRS / INSERM, Paris, France; Sorbonne University, Department of Psychiatry, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Philippe Fossati
- Paris Brain Institute - Institut du Cerveau (ICM), UMR 7225 / UMRS 1127, Sorbonne University / CNRS / INSERM, Paris, France; Sorbonne University, Department of Philosophy, Science Norm Democracy Research Unit, UMR, 8011, Paris, France
| |
Collapse
|
23
|
Gold ND, Mallard AJ, Hermann JC, Zeifman RJ, Pagni BA, Bogenschutz MP, Ross S. Exploring the Potential Utility of Psychedelic Therapy for Patients With Amyotrophic Lateral Sclerosis. J Palliat Med 2023; 26:1408-1418. [PMID: 37167080 DOI: 10.1089/jpm.2022.0604] [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] [Indexed: 05/13/2023] Open
Abstract
Background: Amyotrophic lateral sclerosis (ALS) is an aggressive, terminal neurodegenerative disease that causes death of motor neurons and has an average survival time of 3-4 years. ALS is the most common motor neuron degenerative disease and is increasing in prevalence. There is a pressing need for more effective ALS treatments as available pharmacotherapies do not reverse disease progression or provide substantial clinical benefit. Furthermore, despite psychological distress being highly prevalent in ALS patients, psychological treatments remain understudied. Psychedelics (i.e., serotonergic psychedelics and related compounds like ketamine) have seen a resurgence of research into therapeutic applications for treating a multitude of neuropsychiatric conditions, including psychiatric and existential distress in life-threatening illnesses. Methods: We conducted a narrative review to examine the potential of psychedelic assisted-psychotherapy (PAP) to alleviate psychiatric and psychospiritual distress in ALS. We also discussed the safety of using psychedelics in this population and proposed putative neurobiological mechanisms that may therapeutically intervene on ALS neuropathology. Results: PAP has the potential to treat psychological dimensions and may also intervene on neuropathological dimensions of ALS. Robust improvements in psychiatric and psychospiritual distress from PAP in other populations provide a strong rationale for utilizing this therapy to treat ALS-related psychiatric and existential distress. Furthermore, relevant neuroprotective properties of psychedelics warrant future preclinical trials to investigate this area in ALS models. Conclusion: PAP has the potential to serve as an effective treatment in ALS. Given the lack of effective treatment options, researchers should rigorously explore this therapy for ALS in future trials.
Collapse
Affiliation(s)
- Noah D Gold
- Department of Psychiatry, NYU Langone Center for Psychedelic Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Austin J Mallard
- Department of Psychiatry, NYU Langone Center for Psychedelic Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Jacob C Hermann
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Richard J Zeifman
- Department of Psychiatry, NYU Langone Center for Psychedelic Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Broc A Pagni
- Department of Psychiatry, NYU Langone Center for Psychedelic Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Michael P Bogenschutz
- Department of Psychiatry, NYU Langone Center for Psychedelic Medicine, NYU Grossman School of Medicine, New York, New York, USA
| | - Stephen Ross
- Department of Psychiatry, NYU Langone Center for Psychedelic Medicine, NYU Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
24
|
Priest MF, Freda SN, Rieth IJ, Badong D, Dumrongprechachan V, Kozorovitskiy Y. Peptidergic and functional delineation of the Edinger-Westphal nucleus. Cell Rep 2023; 42:112992. [PMID: 37594894 PMCID: PMC10512657 DOI: 10.1016/j.celrep.2023.112992] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 06/15/2023] [Accepted: 07/31/2023] [Indexed: 08/20/2023] Open
Abstract
Many neuronal populations that release fast-acting excitatory and inhibitory neurotransmitters in the brain also contain slower-acting neuropeptides. These facultative peptidergic cell types are common, but it remains uncertain whether neurons that solely release peptides exist. Our fluorescence in situ hybridization, genetically targeted electron microscopy, and electrophysiological characterization suggest that most neurons of the non-cholinergic, centrally projecting Edinger-Westphal nucleus in mice are obligately peptidergic. We further show, using anterograde projection mapping, monosynaptic retrograde tracing, angled-tip fiber photometry, and chemogenetic modulation and genetically targeted ablation in conjunction with canonical assays for anxiety, that this peptidergic population activates in response to loss of motor control and promotes anxiety responses. Together, these findings elucidate an integrative, ethologically relevant role for the Edinger-Westphal nucleus and functionally align the nucleus with the periaqueductal gray, where it resides. This work advances our understanding of peptidergic modulation of anxiety and provides a framework for future investigations of peptidergic systems.
Collapse
Affiliation(s)
- Michael F Priest
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Sara N Freda
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Isabelle J Rieth
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Deanna Badong
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA
| | - Vasin Dumrongprechachan
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA
| | - Yevgenia Kozorovitskiy
- Department of Neurobiology, Northwestern University, Evanston, IL 60208, USA; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
25
|
Burrows M, Kotoula V, Dipasquale O, Stringaris A, Mehta MA. Ketamine-induced changes in resting state connectivity, 2 h after the drug administration in patients with remitted depression. J Psychopharmacol 2023; 37:784-794. [PMID: 37491833 DOI: 10.1177/02698811231189432] [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] [Indexed: 07/27/2023]
Abstract
BACKGROUND Resting state connectivity studies link ketamine's antidepressant effects with normalisation of the brain connectivity changes that are observed in depression. These changes, however, usually co-occur with improvement in depressive symptoms, making it difficult to attribute these changes to ketamine's effects per se. AIMS Our aim is to examine the effects of ketamine in brain connectivity, 2 h after its administration in a cohort of volunteers with remitted depression. Any significant changes observed in this study could provide insight of ketamine's antidepressant mechanism as they are not accompanied by symptom changes. METHODS In total, 35 participants with remitted depression (21 females, mean age = 28.5 years) participated in a double-blind, placebo-controlled study of ketamine (0.5 mg/kg) or saline. Resting state scans were acquired approximately 2 h after the ketamine infusion. Brain connectivity was examined using a seed-based approach (ventral striatum, amygdala, hippocampus, posterior cingulate cortex and subgenual anterior cingulate cortex (sgACC)) and a brain network analysis (independent component analysis). RESULTS Decreased connectivity between the sgACC and the amygdala was observed approximately 2 h after the ketamine infusion, compared to placebo (pFWE < 0.05). The executive network presented with altered connectivity with different cortical and subcortical regions. Within the network, the left hippocampus and right amygdala had decreased connectivity (pFWE < 0.05). CONCLUSIONS Our findings support a model whereby ketamine would change the connectivity of brain areas and networks that are important for cognitive processing and emotional regulation. These changes could also be an indirect indicator of the plasticity changes induced by the drug.
Collapse
Affiliation(s)
- Matthew Burrows
- Centre for Neuroimaging Sciences, IoPPN, King's College London, London, UK
| | - Vasileia Kotoula
- Experimental Therapeutics and Pathophysiology Branch, NIMH, Bethesda, MA, USA
| | - Ottavia Dipasquale
- Centre for Neuroimaging Sciences, IoPPN, King's College London, London, UK
| | - Argyris Stringaris
- Division of Psychiatry and Department of Clinical, Educational & Health Psychology, UCL, London, UK
- First Department of Psychiatry, National and Kapodistrian University of Athens, Athens, Greece
| | - Mitul A Mehta
- Centre for Neuroimaging Sciences, IoPPN, King's College London, London, UK
| |
Collapse
|
26
|
Krüssel S, Deb I, Son S, Ewall G, Chang M, Lee HK, do Heo W, Kwon HB. Exuberant de novo dendritic spine growth in mature neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.21.550095. [PMID: 37546796 PMCID: PMC10401948 DOI: 10.1101/2023.07.21.550095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Dendritic spines are structural correlates of excitatory synapses maintaining stable synaptic communications. However, this strong spine-synapse relationship was mainly characterized in excitatory pyramidal neurons (PyNs), raising a possibility that inferring synaptic density from dendritic spine number may not be universally applied to all neuronal types. Here we found that the ectopic expression of H-Ras increased dendritic spine numbers regardless of cortical cell types such as layer 2/3 pyramidal neurons (PyNs), parvalbumin (PV)- and vasoactive intestinal peptide (VIP)-positive interneurons (INs) in the primary motor cortex (M1). The probability of detecting dendritic spines was positively correlated with the magnitude of H-Ras activity, suggesting elevated local H-Ras activity is involved in the process of dendritic spine formation. H-Ras overexpression caused high spine turnover rate via adding more spines rather than eliminating them. Two-photon photolysis of glutamate triggered de novo dendritic spine formation in mature neurons, suggesting H-Ras induced spine formation is not restricted to the early development. In PyNs and PV-INs, but not VIP-INs, we observed a shift in average spine neck length towards longer filopodia-like phenotypes. The portion of dendritic spines lacking key excitatory synaptic proteins were significantly increased in H-Ras transfected neurons, suggesting that these increased spines have other distinct functions. High spine density caused by H-Ras did not result in change in the frequency or the amplitude of miniature excitatory postsynaptic currents (mEPSCs). Thus, our results propose that dendritic spines possess more multifaceted functions beyond the morphological proxy of excitatory synapse.
Collapse
Affiliation(s)
- Sarah Krüssel
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ishana Deb
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Seungkyu Son
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Gabrielle Ewall
- Solomon H. Snyder Department of Neuroscience, Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Minhyeok Chang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hey-Kyoung Lee
- Solomon H. Snyder Department of Neuroscience, Zanvyl-Krieger Mind/Brain Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, United States
| | - Won do Heo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Hyung-Bae Kwon
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
27
|
Zhornitsky S, Oliva HNP, Jayne LA, Allsop ASA, Kaye AP, Potenza MN, Angarita GA. Changes in synaptic markers after administration of ketamine or psychedelics: a systematic scoping review. Front Psychiatry 2023; 14:1197890. [PMID: 37435405 PMCID: PMC10331617 DOI: 10.3389/fpsyt.2023.1197890] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/05/2023] [Indexed: 07/13/2023] Open
Abstract
Background Ketamine and psychedelics have abuse liability. They can also induce "transformative experiences" where individuals experience enhanced states of awareness. This enhanced awareness can lead to changes in preexisting behavioral patterns which could be beneficial in the treatment of substance use disorders (SUDs). Preclinical and clinical studies suggest that ketamine and psychedelics may alter markers associated with synaptic density, and that these changes may underlie effects such as sensitization, conditioned place preference, drug self-administration, and verbal memory performance. In this scoping review, we examined studies that measured synaptic markers in animals and humans after exposure to ketamine and/or psychedelics. Methods A systematic search was conducted following PRISMA guidelines, through PubMed, EBSCO, Scopus, and Web of Science, based on a published protocol (Open Science Framework, DOI: 10.17605/OSF.IO/43FQ9). Both in vivo and in vitro studies were included. Studies on the following synaptic markers were included: dendritic structural changes, PSD-95, synapsin-1, synaptophysin-1, synaptotagmin-1, and SV2A. Results Eighty-four studies were included in the final analyses. Seventy-one studies examined synaptic markers following ketamine treatment, nine examined psychedelics, and four examined both. Psychedelics included psilocybin/psilocin, lysergic acid diethylamide, N,N-dimethyltryptamine, 2,5-dimethoxy-4-iodoamphetamine, and ibogaine/noribogaine. Mixed findings regarding synaptic changes in the hippocampus and prefrontal cortex (PFC) have been reported when ketamine was administered in a single dose under basal conditions. Similar mixed findings were seen under basal conditions in studies that used repeated administration of ketamine. However, studies that examined animals during stressful conditions found that a single dose of ketamine counteracted stress-related reductions in synaptic markers in the hippocampus and PFC. Repeated administration of ketamine also counteracted stress effects in the hippocampus. Psychedelics generally increased synaptic markers, but results were more consistently positive for certain agents. Conclusion Ketamine and psychedelics can increase synaptic markers under certain conditions. Heterogeneous findings may relate to methodological differences, agents administered (or different formulations of the same agent), sex, and type of markers. Future studies could address seemingly mixed results by using meta-analytical approaches or study designs that more fully consider individual differences.
Collapse
Affiliation(s)
- Simon Zhornitsky
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, United States
| | - Henrique N. P. Oliva
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, United States
| | - Laura A. Jayne
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, United States
| | - Aza S. A. Allsop
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, United States
| | - Alfred P. Kaye
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Connecticut Mental Health Center, New Haven, CT, United States
- Clinical Neurosciences Division, VA National Center for PTSD, West Haven, CT, United States
| | - Marc N. Potenza
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Connecticut Mental Health Center, New Haven, CT, United States
- Child Study Center, Yale University School of Medicine, New Haven, CT, United States
- Department of Neuroscience, Yale University, New Haven, CT, United States
- Connecticut Council on Problem Gambling, Hartford, CT, United States
- Wu Tsai Institute, Yale University, New Haven, CT, United States
| | - Gustavo A. Angarita
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Clinical Neuroscience Research Unit, Connecticut Mental Health Center, New Haven, CT, United States
| |
Collapse
|
28
|
Sequeira MK, Swanson AM, Kietzman HW, Gourley SL. Cocaine and habit training cause dendritic spine rearrangement in the prelimbic cortex. iScience 2023; 26:106240. [PMID: 37153443 PMCID: PMC10156587 DOI: 10.1016/j.isci.2023.106240] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 01/02/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Successfully navigating dynamic environments requires organisms to learn the consequences of their actions. The prelimbic prefrontal cortex (PL) formulates action-consequence memories and is modulated by addictive drugs like cocaine. We trained mice to obtain food rewards and then unexpectedly withheld reinforcement, triggering new action-consequence memory. New memory was disrupted by cocaine when delivered immediately following non-reinforcement, but not when delayed, suggesting that cocaine disrupted memory consolidation. Cocaine also rapidly inactivated cofilin, a primary regulator of the neuronal actin cytoskeleton. This observation led to the discovery that cocaine also within the time of memory consolidation elevated dendritic spine elimination and blunted spine formation rates on excitatory PL neurons, culminating in thin-type spine attrition. Training drug-naive mice to utilize inflexible response strategies also eliminated thin-type dendritic spines. Thus, cocaine may disrupt action-consequence memory, at least in part, by recapitulating neurobiological sequalae occurring in the formation of inflexible habits.
Collapse
Affiliation(s)
- Michelle K. Sequeira
- Graduate Program in Neuroscience, Emory National Primate Research Center, Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, Emory University, Atlanta, GA 30329, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30329, USA
| | - Andrew M. Swanson
- Graduate Program in Neuroscience, Emory National Primate Research Center, Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, Emory University, Atlanta, GA 30329, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30329, USA
| | - Henry W. Kietzman
- Graduate Program in Neuroscience, Emory National Primate Research Center, Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, Emory University, Atlanta, GA 30329, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30329, USA
| | - Shannon L. Gourley
- Graduate Program in Neuroscience, Emory National Primate Research Center, Departments of Pediatrics and Psychiatry and Behavioral Sciences, Emory University School of Medicine, Emory University, Atlanta, GA 30329, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30329, USA
| |
Collapse
|
29
|
Johnston JN, Kadriu B, Allen J, Gilbert JR, Henter ID, Zarate CA. Ketamine and serotonergic psychedelics: An update on the mechanisms and biosignatures underlying rapid-acting antidepressant treatment. Neuropharmacology 2023; 226:109422. [PMID: 36646310 PMCID: PMC9983360 DOI: 10.1016/j.neuropharm.2023.109422] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/09/2023] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
The discovery of ketamine as a rapid-acting antidepressant spurred significant research to understand its underlying mechanisms of action and to identify other novel compounds that may act similarly. Serotonergic psychedelics (SPs) have shown initial promise in treating depression, though the challenge of conducting randomized controlled trials with SPs and the necessity of long-term clinical observation are important limitations. This review summarizes the similarities and differences between the psychoactive effects associated with both ketamine and SPs and the mechanisms of action of these compounds, with a focus on the monoaminergic, glutamatergic, gamma-aminobutyric acid (GABA)-ergic, opioid, and inflammatory systems. Both molecular and neuroimaging aspects are considered. While their main mechanisms of action differ-SPs increase serotonergic signaling while ketamine is a glutamatergic modulator-evidence suggests that the downstream mechanisms of action of both ketamine and SPs include mechanistic target of rapamycin complex 1 (mTORC1) signaling and downstream GABAA receptor activity. The similarities in downstream mechanisms may explain why ketamine, and potentially SPs, exert rapid-acting antidepressant effects. However, research on SPs is still in its infancy compared to the ongoing research that has been conducted with ketamine. For both therapeutics, issues with regulation and proper controls should be addressed before more widespread implementation. This article is part of the Special Issue on "Ketamine and its Metabolites".
Collapse
Affiliation(s)
- Jenessa N Johnston
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Bashkim Kadriu
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Josh Allen
- The Alfred Centre, Department of Neuroscience, Monash University, Melbourne, Victoria, Australia.
| | - Jessica R Gilbert
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Ioline D Henter
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| | - Carlos A Zarate
- Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
30
|
Nguyen TML, Defaix C, Mendez-David I, Tritschler L, Etting I, Alvarez JC, Choucha W, Colle R, Corruble E, David DJ, Gardier AM. Intranasal (R, S)-ketamine delivery induces sustained antidepressant effects associated with changes in cortical balance of excitatory/inhibitory synaptic activity. Neuropharmacology 2023; 225:109357. [PMID: 36462636 DOI: 10.1016/j.neuropharm.2022.109357] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/23/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022]
Abstract
In 2019, an intranasal (IN) spray of esketamine SPRAVATO® was approved as a fast-acting antidepressant by drug Agencies US FDA and European EMA. At sub-anesthetic doses, (±)-ketamine, a non-competitive glutamate N-methyl-d-aspartate (NMDA) receptor antagonist, increases the overall excitability of the medial prefrontal cortex (mPFC), an effect being essential for its rapid antidepressant activity. We wondered if this effect of ketamine could come from changes in the balance between neuronal excitation and inhibition (E/I balance) in the mPFC. Here, we performed a preclinical approach to study neurochemical and behavioral responses to a single IN ketamine dose in BALB/cJ mice, a strain more sensitive to stress. By using in vivo microdialysis, we measured cortical E/I balance as the ratio between glutamate to GABA extracellular levels 24 h post-ketamine. We found, for the first time, that E/I balance was shifted in favor of excitation rather than inhibition in the mPFC but more robustly with IN KET than with a single intraperitoneal (IP) dose. Increases in plasma and brain ketamine, norketamine and HNKs levels suggest different metabolic profiles of IP and IN ketamine 30 min post-dose. A significantly larger proportion of ketamine and HNKs in the brain are derived from the IN route 30 min post-dose. It may be linked to the greater magnitude in E/I ratio following IN delivery relative to IP at t24 h. This study suggests that both IP and IN are effective brain delivery methods inducing similar sustained antidepressant efficacy of KET, but the way they induced neurotransmitter changes is slightly different.
Collapse
Affiliation(s)
- Thi Mai Loan Nguyen
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018, CESP-Inserm, MOODS Team, Chatenay-Malabry, 92290, France
| | - Céline Defaix
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018, CESP-Inserm, MOODS Team, Chatenay-Malabry, 92290, France
| | - Indira Mendez-David
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018, CESP-Inserm, MOODS Team, Chatenay-Malabry, 92290, France
| | - Laurent Tritschler
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018, CESP-Inserm, MOODS Team, Chatenay-Malabry, 92290, France
| | - Isabelle Etting
- Lab. Pharmacologie-Toxicologie, Centre Hospitalier Universitaire Raymond Poincaré, AP-HP, Garches, France
| | - Jean-Claude Alvarez
- Lab. Pharmacologie-Toxicologie, Centre Hospitalier Universitaire Raymond Poincaré, AP-HP, Garches, France
| | - Walid Choucha
- Université Paris-Saclay, Faculté de Médecine, UMR 1018, CESP MOODS Team, Bicêtre Hospital, Université Paris-Saclay, Le Kremin-Bicêtre, 94270, France; Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Hôpital de Bicêtre, Le Kremlin Bicêtre, F-94275, France
| | - Romain Colle
- Université Paris-Saclay, Faculté de Médecine, UMR 1018, CESP MOODS Team, Bicêtre Hospital, Université Paris-Saclay, Le Kremin-Bicêtre, 94270, France; Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Hôpital de Bicêtre, Le Kremlin Bicêtre, F-94275, France
| | - Emmanuelle Corruble
- Université Paris-Saclay, Faculté de Médecine, UMR 1018, CESP MOODS Team, Bicêtre Hospital, Université Paris-Saclay, Le Kremin-Bicêtre, 94270, France; Service Hospitalo-Universitaire de Psychiatrie de Bicêtre, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris-Saclay, Hôpital de Bicêtre, Le Kremlin Bicêtre, F-94275, France
| | - Denis J David
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018, CESP-Inserm, MOODS Team, Chatenay-Malabry, 92290, France
| | - Alain M Gardier
- Université Paris-Saclay, Faculté de Pharmacie, UMR 1018, CESP-Inserm, MOODS Team, Chatenay-Malabry, 92290, France.
| |
Collapse
|
31
|
Sippy T, Tritsch NX. Unraveling the dynamics of dopamine release and its actions on target cells. Trends Neurosci 2023; 46:228-239. [PMID: 36635111 PMCID: PMC10204099 DOI: 10.1016/j.tins.2022.12.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/22/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023]
Abstract
The neuromodulator dopamine (DA) is essential for regulating learning, motivation, and movement. Despite its importance, however, the mechanisms by which DA influences the activity of target cells to alter behavior remain poorly understood. In this review, we describe recent methodological advances that are helping to overcome challenges that have historically hindered the field. We discuss how the employment of these methods is shedding light on the complex dynamics of extracellular DA in the brain, as well as how DA signaling alters the electrical, biochemical, and population activity of target neurons in vivo. These developments are generating novel hypotheses about the mechanisms through which DA release modifies behavior.
Collapse
Affiliation(s)
- Tanya Sippy
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.
| | - Nicolas X Tritsch
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA; Fresco Institute for Parkinson's and Movement Disorders, New York University Langone Health, New York, NY, USA.
| |
Collapse
|
32
|
Davoudian PA, Shao LX, Kwan AC. Shared and Distinct Brain Regions Targeted for Immediate Early Gene Expression by Ketamine and Psilocybin. ACS Chem Neurosci 2023; 14:468-480. [PMID: 36630309 PMCID: PMC9898239 DOI: 10.1021/acschemneuro.2c00637] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Psilocybin is a psychedelic with therapeutic potential. While there is growing evidence that psilocybin exerts its beneficial effects through enhancing neural plasticity, the exact brain regions involved are not completely understood. Determining the impact of psilocybin on plasticity-related gene expression throughout the brain can broaden our understanding of the neural circuits involved in psychedelic-evoked neural plasticity. In this study, whole-brain serial two-photon microscopy and light sheet microscopy were employed to map the expression of the immediate early gene, c-Fos, in male and female mice. The drug-induced c-Fos expression following psilocybin administration was compared to that of subanesthetic ketamine and saline control. Psilocybin and ketamine produced acutely comparable elevations in c-Fos expression in numerous brain regions, including anterior cingulate cortex, locus coeruleus, primary visual cortex, central and basolateral amygdala, medial and lateral habenula, and claustrum. Select regions exhibited drug-preferential differences, such as dorsal raphe and insular cortex for psilocybin and the CA1 subfield of hippocampus for ketamine. To gain insights into the contributions of receptors and cell types, the c-Fos expression maps were related to brain-wide in situ hybridization data. The transcript analyses showed that the endogenous levels of Grin2a and Grin2b predict whether a cortical region is sensitive to drug-evoked neural plasticity for both ketamine and psilocybin. Collectively, the systematic mapping approach produced an unbiased list of brain regions impacted by psilocybin and ketamine. The data are a resource that highlights previously underappreciated regions for future investigations. Furthermore, the robust relationships between drug-evoked c-Fos expression and endogenous transcript distributions suggest glutamatergic receptors as a potential convergent target for how psilocybin and ketamine produce their rapid-acting and long-lasting therapeutic effects.
Collapse
Affiliation(s)
- Pasha A. Davoudian
- Medical Scientist Training Program, Yale University School of Medicine, New Haven, Connecticut, 06511, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, Connecticut, 06511, USA
| | - Ling-Xiao Shao
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, 06511, USA
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, 14853, USA
| | - Alex C. Kwan
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, 06511, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, 06511, USA
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, 14853, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, New York, 10065, USA
| |
Collapse
|
33
|
Increasing Adiponectin Signaling by Sub-Chronic AdipoRon Treatment Elicits Antidepressant- and Anxiolytic-Like Effects Independent of Changes in Hippocampal Plasticity. Biomedicines 2023; 11:biomedicines11020249. [PMID: 36830788 PMCID: PMC9953351 DOI: 10.3390/biomedicines11020249] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/16/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
(1) Background: Adiponectin is an adipocyte-secreted hormone that has antidepressant- and anxiolytic-like effects in preclinical studies. Here, we investigated the antidepressant- and anxiolytic-like effects of sub-chronic treatment with AdipoRon, an adiponectin receptor agonist, and its potential linkage to changes in hippocampal adult neurogenesis and synaptic plasticity. (2) Methods: Different cohorts of wild-type C57BL/6J and CamKIIα-Cre male mice were treated with sub-chronic (7 days) AdipoRon, followed by behavioral, molecular, and electrophysiological experiments. (3) Results: 7-day AdipoRon treatment elicited antidepressant- and anxiolytic-like effects but did not affect hippocampal neurogenesis. AdipoRon treatment reduced hippocampal brain-derived neurotrophic factor (BDNF) levels, neuronal activation in the ventral dentate gyrus, and long-term potentiation of the perforant path. The knockdown of N-methyl-D-aspartate (NMDA) receptor subunits GluN2A and GluN2B in the ventral hippocampus did not affect the antidepressant- and anxiolytic-like effects of AdipoRon. (4) Conclusions: Increasing adiponectin signaling through sub-chronic AdipoRon treatment results in antidepressant- and anxiolytic-like effects independent of changes in hippocampal structural and synaptic function.
Collapse
|
34
|
Marguilho M, Figueiredo I, Castro-Rodrigues P. A unified model of ketamine's dissociative and psychedelic properties. J Psychopharmacol 2023; 37:14-32. [PMID: 36527355 PMCID: PMC9834329 DOI: 10.1177/02698811221140011] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ketamine is an N-methyl-d-aspartate antagonist which is increasingly being researched and used as a treatment for depression. In low doses, it can cause a transitory modification in consciousness which was classically labelled as 'dissociation'. However, ketamine is also commonly classified as an atypical psychedelic and it has been recently reported that ego dissolution experiences during ketamine administration are associated with greater antidepressant response. Neuroimaging studies have highlighted several similarities between the effects of ketamine and those of serotonergic psychedelics in the brain; however, no unified account has been proposed for ketamine's multi-level effects - from molecular to network and psychological levels. Here, we propose that the fast, albeit transient, antidepressant effects observed after ketamine infusions are mainly driven by its acute modulation of reward circuits and sub-acute increase in neuroplasticity, while its dissociative and psychedelic properties are driven by dose- and context-dependent disruption of large-scale functional networks. Computationally, as nodes of the salience network (SN) represent high-level priors about the body ('minimal' self) and nodes of the default-mode network (DMN) represent the highest-level priors about narrative self-experience ('biographical' self), we propose that transitory SN desegregation and disintegration accounts for ketamine's 'dissociative' state, while transitory DMN desegregation and disintegration accounts for ketamine's 'psychedelic' state. In psychedelic-assisted psychotherapy, a relaxation of the highest-level beliefs with psychotherapeutic support may allow a revision of pathological self-representation models, for which neuroplasticity plays a permissive role. Our account provides a multi-level rationale for using the psychedelic properties of ketamine to increase its long-term benefits.
Collapse
Affiliation(s)
| | | | - Pedro Castro-Rodrigues
- Centro Hospitalar Psiquiátrico de Lisboa, Lisbon, Portugal,NOVA Medical School, NMS, Universidade Nova de Lisboa, Lisbon, Portugal,Pedro Castro-Rodrigues, Centro Hospitalar Psiquiátrico de Lisboa, Avenida do Brasil, 53, Lisbon, 1749-002, Portugal.
| |
Collapse
|
35
|
Calder AE, Hasler G. Towards an understanding of psychedelic-induced neuroplasticity. Neuropsychopharmacology 2023; 48:104-112. [PMID: 36123427 PMCID: PMC9700802 DOI: 10.1038/s41386-022-01389-z] [Citation(s) in RCA: 114] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 12/20/2022]
Abstract
Classic psychedelics, such as LSD, psilocybin, and the DMT-containing beverage ayahuasca, show some potential to treat depression, anxiety, and addiction. Importantly, clinical improvements can last for months or years after treatment. It has been theorized that these long-term improvements arise because psychedelics rapidly and lastingly stimulate neuroplasticity. The focus of this review is on answering specific questions about the effects of psychedelics on neuroplasticity. Firstly, we review the evidence that psychedelics promote neuroplasticity and examine the cellular and molecular mechanisms behind the effects of different psychedelics on different aspects of neuroplasticity, including dendritogenesis, synaptogenesis, neurogenesis, and expression of plasticity-related genes (e.g., brain-derived neurotrophic factor and immediate early genes). We then examine where in the brain psychedelics promote neuroplasticity, particularly discussing the prefrontal cortex and hippocampus. We also examine what doses are required to produce this effect (e.g., hallucinogenic doses vs. "microdoses"), and how long purported changes in neuroplasticity last. Finally, we discuss the likely consequences of psychedelics' effects on neuroplasticity for both patients and healthy people, and we identify important research questions that would further scientific understanding of psychedelics' effects on neuroplasticity and its potential clinical applications.
Collapse
Affiliation(s)
- Abigail E Calder
- University Center for Psychiatric Research, University of Fribourg, Fribourg, Switzerland.
| | - Gregor Hasler
- University Center for Psychiatric Research, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
36
|
Obray JD, Landin JD, Vaughan DT, Scofield MD, Chandler LJ. Adolescent alcohol exposure reduces dopamine 1 receptor modulation of prelimbic neurons projecting to the nucleus accumbens and basolateral amygdala. ADDICTION NEUROSCIENCE 2022; 4:100044. [PMID: 36643604 PMCID: PMC9836047 DOI: 10.1016/j.addicn.2022.100044] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Binge drinking during adolescence is highly prevalent despite increasing evidence of its long-term impact on behaviors associated with modulation of behavioral flexibility by the medial prefrontal cortex (mPFC). In the present study, male and female rats underwent adolescent intermittent ethanol (AIE) exposure by vapor inhalation. After aging to adulthood, retrograde bead labelling and viral tagging were used to identify populations of neurons in the prelimbic region (PrL) of the mPFC that project to specific subcortical targets. Electrophysiological recording from bead-labelled neurons in PrL slices revealed that AIE did not alter the intrinsic excitability of PrL neurons that projected to either the NAc or the BLA. Similarly, recordings of spontaneous inhibitory and excitatory post-synaptic currents revealed no AIE-induced changes in synaptic drive onto either population of projection neurons. In contrast, AIE exposure was associated with a loss of dopamine receptor 1 (D1), but no change in dopamine receptor 2 (D2), modulation of evoked firing of both populations of projection neurons. Lastly, confocal imaging of proximal and apical dendritic tufts of viral-labelled PrL neurons that projected to the nucleus accumbens (NAc) revealed AIE did not alter the density of dendritic spines. Together, these observations provide evidence that AIE exposure results in disruption of D1 receptor modulation of PrL inputs to at least two major subcortical target regions that have been implicated in AIE-induced long-term changes in behavioral control.
Collapse
Affiliation(s)
- J. Daniel Obray
- Department of Neuroscience, Medical University of South Carolina, 30 Courtenay Drive, Charleston SC 29425, USA
| | - Justine D. Landin
- Department of Neuroscience, Medical University of South Carolina, 30 Courtenay Drive, Charleston SC 29425, USA
| | - Dylan T. Vaughan
- Department of Neuroscience, Medical University of South Carolina, 30 Courtenay Drive, Charleston SC 29425, USA
| | - Michael D. Scofield
- Department of Neuroscience, Medical University of South Carolina, 30 Courtenay Drive, Charleston SC 29425, USA,Department of Anesthesiology, Medical University of South Carolina, Charleston SC, USA
| | - L. Judson Chandler
- Department of Neuroscience, Medical University of South Carolina, 30 Courtenay Drive, Charleston SC 29425, USA,Corresponding author. (L.J. Chandler)
| |
Collapse
|
37
|
Wu Y, Wu M, Vázquez-Guardado A, Kim J, Zhang X, Avila R, Kim JT, Deng Y, Yu Y, Melzer S, Bai Y, Yoon H, Meng L, Zhang Y, Guo H, Hong L, Kanatzidis EE, Haney CR, Waters EA, Banks AR, Hu Z, Lie F, Chamorro LP, Sabatini BL, Huang Y, Kozorovitskiy Y, Rogers JA. Wireless multi-lateral optofluidic microsystems for real-time programmable optogenetics and photopharmacology. Nat Commun 2022; 13:5571. [PMID: 36137999 PMCID: PMC9500026 DOI: 10.1038/s41467-022-32947-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 08/24/2022] [Indexed: 11/27/2022] Open
Abstract
In vivo optogenetics and photopharmacology are two techniques for controlling neuronal activity that have immense potential in neuroscience research. Their applications in tether-free groups of animals have been limited in part due to tools availability. Here, we present a wireless, battery-free, programable multilateral optofluidic platform with user-selected modalities for optogenetics, pharmacology and photopharmacology. This system features mechanically compliant microfluidic and electronic interconnects, capabilities for dynamic control over the rates of drug delivery and real-time programmability, simultaneously for up to 256 separate devices in a single cage environment. Our behavioral experiments demonstrate control of motor behaviors in grouped mice through in vivo optogenetics with co-located gene delivery and controlled photolysis of caged glutamate. These optofluidic systems may expand the scope of wireless techniques to study neural processing in animal models.
Collapse
Affiliation(s)
- Yixin Wu
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
| | - Mingzheng Wu
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Abraham Vázquez-Guardado
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
| | - Joohee Kim
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Center for Bionics of Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Xin Zhang
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Raudel Avila
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
| | - Jin-Tae Kim
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
| | - Yujun Deng
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
- State Key Laboratory of Mechanical System and Vibration, Shanghai Jiao Tong University, Shanghai, China
| | | | - Sarah Melzer
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Ave, Boston, MA, 02115, USA
| | - Yun Bai
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
| | - Hyoseo Yoon
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Lingzi Meng
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
| | - Yi Zhang
- Polymer Program, Institute of Materials Science, University of Connecticut, Storrs, CT, 06269, US
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, US
| | - Hexia Guo
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
| | - Liu Hong
- Mechanical Science and Engineering Department, University of Illinois, Urbana, IL, USA
| | - Evangelos E Kanatzidis
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Chad R Haney
- Center for Advanced Molecular Imaging, Radiology, and Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Emily A Waters
- Center for Advanced Molecular Imaging, Radiology, and Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Anthony R Banks
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Neurolux Inc, Northfield, IL, USA
| | - Ziying Hu
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
| | | | - Leonardo P Chamorro
- Mechanical Science and Engineering Department, University of Illinois, Urbana, IL, USA
| | - Bernardo L Sabatini
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Ave, Boston, MA, 02115, USA
| | - Yonggang Huang
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA.
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA.
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, USA.
| | - Yevgenia Kozorovitskiy
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.
- Chemistry of Life Processes Institutes, Northwestern University, Evanston, IL, USA.
| | - John A Rogers
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA.
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA.
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA.
- Neurolux Inc, Northfield, IL, USA.
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Simpson Querrey Institute & Feinberg Medical School, Northwestern University, Evanston, IL, USA.
- Department of Chemistry, Northwestern University, Evanston, IL, USA.
- Department of Neurological Surgery, Northwestern University, Evanston, IL, USA.
- Department of Electrical and Computer Engineering, Northwestern University, Evanston, IL, USA.
- Department of Computer Science, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
38
|
Tian H, Hu Z, Xu J, Wang C. The molecular pathophysiology of depression and the new therapeutics. MedComm (Beijing) 2022; 3:e156. [PMID: 35875370 PMCID: PMC9301929 DOI: 10.1002/mco2.156] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/06/2022] [Accepted: 06/06/2022] [Indexed: 12/21/2022] Open
Abstract
Major depressive disorder (MDD) is a highly prevalent and disabling disorder. Despite the many hypotheses proposed to understand the molecular pathophysiology of depression, it is still unclear. Current treatments for depression are inadequate for many individuals, because of limited effectiveness, delayed efficacy (usually two weeks), and side effects. Consequently, novel drugs with increased speed of action and effectiveness are required. Ketamine has shown to have rapid, reliable, and long-lasting antidepressant effects in treatment-resistant MDD patients and represent a breakthrough therapy for patients with MDD; however, concerns regarding its efficacy, potential misuse, and side effects remain. In this review, we aimed to summarize molecular mechanisms and pharmacological treatments for depression. We focused on the fast antidepressant treatment and clarified the safety, tolerability, and efficacy of ketamine and its metabolites for the MDD treatment, along with a review of the potential pharmacological mechanisms, research challenges, and future clinical prospects.
Collapse
Affiliation(s)
- Haihua Tian
- Ningbo Key Laboratory of Behavioral NeuroscienceNingbo University School of MedicineNingboZhejiangChina
- Zhejiang Provincial Key Laboratory of PathophysiologySchool of MedicineNingbo UniversityNingboZhejiangChina
- Department of Physiology and PharmacologyNingbo University School of MedicineNingboZhejiangChina
- Department of Laboratory MedicineNingbo Kangning HospitalNingboZhejiangChina
| | - Zhenyu Hu
- Department of Child PsychiatryNingbo Kanning HospitalNingboZhejiangChina
| | - Jia Xu
- Ningbo Key Laboratory of Behavioral NeuroscienceNingbo University School of MedicineNingboZhejiangChina
- Zhejiang Provincial Key Laboratory of PathophysiologySchool of MedicineNingbo UniversityNingboZhejiangChina
- Department of Physiology and PharmacologyNingbo University School of MedicineNingboZhejiangChina
| | - Chuang Wang
- Ningbo Key Laboratory of Behavioral NeuroscienceNingbo University School of MedicineNingboZhejiangChina
- Zhejiang Provincial Key Laboratory of PathophysiologySchool of MedicineNingbo UniversityNingboZhejiangChina
- Department of Physiology and PharmacologyNingbo University School of MedicineNingboZhejiangChina
| |
Collapse
|
39
|
Rogeau A, Nordio G, Veronese M, Brown K, Nour MM, Osugo M, Jauhar S, Howes OD, McCutcheon RA. The relationship between glutamate, dopamine, and cortical gray matter: A simultaneous PET-MR study. Mol Psychiatry 2022; 27:3493-3500. [PMID: 35546633 PMCID: PMC9708555 DOI: 10.1038/s41380-022-01596-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 04/14/2022] [Accepted: 04/21/2022] [Indexed: 11/09/2022]
Abstract
Prefrontal cortex has been shown to regulate striatal dopaminergic function via glutamatergic mechanisms in preclinical studies. Concurrent disruption of these systems is also often seen in neuropsychiatric disease. The simultaneous measurement of striatal dopamine signaling, cortical gray matter, and glutamate levels is therefore of major interest, but has not been previously reported. In the current study, twenty-eight healthy subjects underwent 2 simultaneous [11C]-( + )-PHNO PET-MRI scans, once after placebo and once after amphetamine in a double-blind randomized cross-over design, to measure striatal dopamine release, striatal dopamine receptor (D2/3R) availability, anterior cingulate glutamate+glutamine (Glx) levels, and cortical gray matter volumes at the same time. Voxel-based morphometry was used to investigate associations between neurochemical measures and gray matter volumes. Whole striatum D2/3R availability was positively associated with prefrontal cortex gray matter volume (pFWE corrected = 0.048). This relationship was mainly driven by associative receptor availability (pFWE corrected = 0.023). In addition, an interaction effect was observed between sensorimotor striatum D2/3R availability and anterior cingulate Glx, such that in individuals with greater anterior cingulate Glx concentrations, D2/3R availability was negatively associated with right frontal cortex gray matter volumes, while a positive D2/3R-gray matter association was observed in individuals with lower anterior cingulate Glx levels (pFWE corrected = 0.047). These results are consistent with the hypothesis that the prefrontal cortex is involved in regulation of striatal dopamine function. Furthermore, the observed associations raise the possibility that this regulation may be modulated by anterior cingulate glutamate concentrations.
Collapse
Affiliation(s)
- Antoine Rogeau
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Department of Nuclear Medicine, Lille University Hospitals, Lille, France
| | - Giovanna Nordio
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Department of Information Engineering, University of Padua, Padua, Italy
| | - Kirsten Brown
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Matthew M Nour
- Max Planck UCL Centre for Computational Psychiatry and Ageing Research, University College London, London, UK
| | - Martin Osugo
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Hammersmith Hospital, Imperial College London, London, UK
| | - Sameer Jauhar
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Hammersmith Hospital, Imperial College London, London, UK
| | - Oliver D Howes
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Hammersmith Hospital, Imperial College London, London, UK
| | - Robert A McCutcheon
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK.
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Hammersmith Hospital, Imperial College London, London, UK.
| |
Collapse
|
40
|
Abstract
Depression is an episodic form of mental illness characterized by mood state transitions with poorly understood neurobiological mechanisms. Antidepressants reverse the effects of stress and depression on synapse function, enhancing neurotransmission, increasing plasticity, and generating new synapses in stress-sensitive brain regions. These properties are shared to varying degrees by all known antidepressants, suggesting that synaptic remodeling could play a key role in depression pathophysiology and antidepressant function. Still, it is unclear whether and precisely how synaptogenesis contributes to mood state transitions. Here, we review evidence supporting an emerging model in which depression is defined by a distinct brain state distributed across multiple stress-sensitive circuits, with neurons assuming altered functional properties, synapse configurations, and, importantly, a reduced capacity for plasticity and adaptation. Antidepressants act initially by facilitating plasticity and enabling a functional reconfiguration of this brain state. Subsequently, synaptogenesis plays a specific role in sustaining these changes over time.
Collapse
Affiliation(s)
- Puja K Parekh
- Department of Psychiatry and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA;
| | - Shane B Johnson
- Department of Psychiatry and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA;
| | - Conor Liston
- Department of Psychiatry and Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York, USA;
| |
Collapse
|
41
|
Xu S, Yao X, Li B, Cui R, Zhu C, Wang Y, Yang W. Uncovering the Underlying Mechanisms of Ketamine as a Novel Antidepressant. Front Pharmacol 2022; 12:740996. [PMID: 35872836 PMCID: PMC9301111 DOI: 10.3389/fphar.2021.740996] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/20/2021] [Indexed: 12/26/2022] Open
Abstract
Major depressive disorder (MDD) is a devastating psychiatric disorder which exacts enormous personal and social-economic burdens. Ketamine, an N-methyl-D-aspartate receptor (NMDAR) antagonist, has been discovered to exert rapid and sustained antidepressant-like actions on MDD patients and animal models. However, the dissociation and psychotomimetic propensities of ketamine have limited its use for psychiatric indications. Here, we review recently proposed mechanistic hypotheses regarding how ketamine exerts antidepressant-like actions. Ketamine may potentiate α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptor (AMPAR)-mediated transmission in pyramidal neurons by disinhibition and/or blockade of spontaneous NMDAR-mediated neurotransmission. Ketamine may also activate neuroplasticity- and synaptogenesis-relevant signaling pathways, which may converge on key components like brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB) and mechanistic target of rapamycin (mTOR). These processes may subsequently rebalance the excitatory/inhibitory transmission and restore neural network integrity that is compromised in depression. Understanding the mechanisms underpinning ketamine’s antidepressant-like actions at cellular and neural circuit level will drive the development of safe and effective pharmacological interventions for the treatment of MDD.
Collapse
Affiliation(s)
- Songbai Xu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, China
| | - Xiaoxiao Yao
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Cuilin Zhu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Cuilin Zhu, ; Yao Wang, ; Wei Yang,
| | - Yao Wang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Cuilin Zhu, ; Yao Wang, ; Wei Yang,
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Cuilin Zhu, ; Yao Wang, ; Wei Yang,
| |
Collapse
|
42
|
Burlingham SR, Wong NF, Peterkin L, Lubow L, Dos Santos Passos C, Benner O, Ghebrial M, Cast TP, Xu-Friedman MA, Südhof TC, Chanda S. Induction of synapse formation by de novo neurotransmitter synthesis. Nat Commun 2022; 13:3060. [PMID: 35650274 PMCID: PMC9160008 DOI: 10.1038/s41467-022-30756-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 05/17/2022] [Indexed: 11/09/2022] Open
Abstract
A vital question in neuroscience is how neurons align their postsynaptic structures with presynaptic release sites. Although synaptic adhesion proteins are known to contribute in this process, the role of neurotransmitters remains unclear. Here we inquire whether de novo biosynthesis and vesicular release of a noncanonical transmitter can facilitate the assembly of its corresponding postsynapses. We demonstrate that, in both stem cell-derived human neurons as well as in vivo mouse neurons of purely glutamatergic identity, ectopic expression of GABA-synthesis enzymes and vesicular transporters is sufficient to both produce GABA from ambient glutamate and transmit it from presynaptic terminals. This enables efficient accumulation and consistent activation of postsynaptic GABAA receptors, and generates fully functional GABAergic synapses that operate in parallel but independently of their glutamatergic counterparts. These findings suggest that presynaptic release of a neurotransmitter itself can signal the organization of relevant postsynaptic apparatus, which could be directly modified to reprogram the synapse identity of neurons.
Collapse
Affiliation(s)
- Scott R Burlingham
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Nicole F Wong
- Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Lindsay Peterkin
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Lily Lubow
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | | | - Orion Benner
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | - Michael Ghebrial
- Biological Science, California State University Fullerton, Fullerton, CA, USA
| | - Thomas P Cast
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, USA
| | | | - Thomas C Südhof
- Molecular & Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Soham Chanda
- Biochemistry & Molecular Biology, Colorado State University, Fort Collins, CO, USA.
- Molecular & Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
- Molecular, Cellular & Integrated Neurosciences, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
43
|
Wilke SA, Lavi K, Byeon S, Donohue KC, Sohal VS. Convergence of Clinically Relevant Manipulations on Dopamine-Regulated Prefrontal Activity Underlying Stress Coping Responses. Biol Psychiatry 2022; 91:810-820. [PMID: 35090617 PMCID: PMC11182612 DOI: 10.1016/j.biopsych.2021.11.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Depression is pleiotropic and influenced by diverse genetic, environmental, and pharmacological factors. Identifying patterns of circuit activity on which many of these factors converge would be important, because studying these patterns could reveal underlying pathophysiological processes and/or novel therapies. Depression is commonly assumed to involve changes within prefrontal circuits, and dopamine D2 receptor (D2R) agonists are increasingly used as adjunctive antidepressants. Nevertheless, how D2Rs influence disease-relevant patterns of prefrontal circuit activity remains unknown. METHODS We used brain slice calcium imaging to measure how patterns of prefrontal activity are modulated by D2Rs, antidepressants, and manipulations that increase depression susceptibility. To validate the idea that prefrontal D2Rs might contribute to antidepressant responses, we used optogenetic and genetic manipulations to test how dopamine, D2Rs, and D2R+ neurons contribute to stress-coping behavior. RESULTS Patterns of positively correlated activity in prefrontal microcircuits are specifically enhanced by D2R stimulation as well as by two mechanistically distinct antidepressants, ketamine and fluoxetine. Conversely, this D2R-driven effect was disrupted in two etiologically distinct depression models, a genetic susceptibility model and mice that are susceptible to chronic social defeat. Phasic stimulation of dopaminergic afferents to the prefrontal cortex and closed-loop stimulation of D2R+ neurons increased effortful responses to tail suspension stress, whereas prefrontal D2R deletion reduced the duration of individual struggling episodes. CONCLUSIONS Correlated prefrontal microcircuit activity represents a point of convergence for multiple depression-related manipulations. Prefrontal D2Rs enhance this activity. Through this mechanism, prefrontal D2Rs may promote network states associated with antidepressant actions and effortful responses to stress.
Collapse
Affiliation(s)
- Scott A Wilke
- Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; Department of Psychiatry and Behavior Sciences, University of California, San Francisco, San Francisco, California; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, California
| | - Karen Lavi
- Department of Psychiatry and Behavior Sciences, University of California, San Francisco, San Francisco, California; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, California
| | - Sujin Byeon
- Department of Psychiatry and Behavior Sciences, University of California, San Francisco, San Francisco, California; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, California
| | - Kevin C Donohue
- Department of Psychiatry and Behavior Sciences, University of California, San Francisco, San Francisco, California; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, California
| | - Vikaas S Sohal
- Department of Psychiatry and Behavior Sciences, University of California, San Francisco, San Francisco, California; Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, California; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, California.
| |
Collapse
|
44
|
Canton-Josh JE, Qin J, Salvo J, Kozorovitskiy Y. Dopaminergic regulation of vestibulo-cerebellar circuits through unipolar brush cells. eLife 2022; 11:e76912. [PMID: 35476632 PMCID: PMC9106328 DOI: 10.7554/elife.76912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
While multiple monoamines modulate cerebellar output, the mechanistic details of dopaminergic signaling in the cerebellum remain poorly understood. We show that dopamine type 1 receptors (Drd1) are expressed in unipolar brush cells (UBCs) of the mouse cerebellar vermis. Drd1 activation increases UBC firing rate and post-synaptic NMDAR -mediated currents. Using anatomical tracing and in situ hybridization, we test three hypotheses about the source of cerebellar dopamine. We exclude midbrain dopaminergic nuclei and tyrosine hydroxylase-positive Purkinje (Pkj) cells as potential sources, supporting the possibility of dopaminergic co-release from locus coeruleus (LC) axons. Using an optical dopamine sensor GRABDA2h, electrical stimulation, and optogenetic activation of LC fibers in the acute slice, we find evidence for monoamine release onto Drd1-expressing UBCs. Altogether, we propose that the LC regulates cerebellar cortex activity by co-releasing dopamine onto UBCs to modulate their response to cerebellar inputs. Pkj cells directly inhibit these Drd1-positive UBCs, forming a dopamine-sensitive recurrent vestibulo-cerebellar circuit.
Collapse
Affiliation(s)
| | - Joanna Qin
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
| | - Joseph Salvo
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
| | | |
Collapse
|
45
|
Sala N, Paoli C, Bonifacino T, Mingardi J, Schiavon E, La Via L, Milanese M, Tornese P, Datusalia AK, Rosa J, Facchinetti R, Frumento G, Carini G, Salerno Scarzella F, Scuderi C, Forti L, Barbon A, Bonanno G, Popoli M, Musazzi L. Acute Ketamine Facilitates Fear Memory Extinction in a Rat Model of PTSD Along With Restoring Glutamatergic Alterations and Dendritic Atrophy in the Prefrontal Cortex. Front Pharmacol 2022; 13:759626. [PMID: 35370690 PMCID: PMC8968915 DOI: 10.3389/fphar.2022.759626] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 01/25/2022] [Indexed: 12/17/2022] Open
Abstract
Stress represents a major risk factor for psychiatric disorders, including post-traumatic stress disorder (PTSD). Recently, we dissected the destabilizing effects of acute stress on the excitatory glutamate system in the prefrontal cortex (PFC). Here, we assessed the effects of single subanesthetic administration of ketamine (10 mg/kg) on glutamate transmission and dendritic arborization in the PFC of footshock (FS)-stressed rats, along with changes in depressive, anxious, and fear extinction behaviors. We found that ketamine, while inducing a mild increase of glutamate release in the PFC of naïve rats, blocked the acute stress-induced enhancement of glutamate release when administered 24 or 72 h before or 6 h after FS. Accordingly, the treatment with ketamine 6 h after FS also reduced the stress-dependent increase of spontaneous excitatory postsynaptic current (sEPSC) amplitude in prelimbic (PL)-PFC. At the same time, ketamine injection 6 h after FS was found to rescue apical dendritic retraction of pyramidal neurons induced by acute stress in PL-PFC and facilitated contextual fear extinction. These results show rapid effects of ketamine in animals subjected to acute FS, in line with previous studies suggesting a therapeutic action of the drug in PTSD models. Our data are consistent with a mechanism of ketamine involving re-establishment of synaptic homeostasis, through restoration of glutamate release, and structural remodeling of dendrites.
Collapse
Affiliation(s)
- Nathalie Sala
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy
| | - Caterina Paoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy.,School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Tiziana Bonifacino
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Jessica Mingardi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Emanuele Schiavon
- Department of Biotechnology and Life Sciences, University of Insubria, Busto Arsizio, Italy
| | - Luca La Via
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marco Milanese
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Paolo Tornese
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy
| | - Ashok K Datusalia
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy.,Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, India
| | - Jessica Rosa
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy.,Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirao Preto, Brazil
| | - Roberta Facchinetti
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy
| | - Giulia Frumento
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy
| | - Giulia Carini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | | | - Caterina Scuderi
- Department of Physiology and Pharmacology "Vittorio Erspamer", SAPIENZA University of Rome, Rome, Italy
| | - Lia Forti
- Department of Biotechnology and Life Sciences, University of Insubria, Busto Arsizio, Italy
| | - Alessandro Barbon
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giambattista Bonanno
- Department of Pharmacy, Unit of Pharmacology and Toxicology, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Maurizio Popoli
- Laboratory of Neuropsychopharmacology and Functional Neurogenomics, Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Milano, Italy
| | - Laura Musazzi
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
46
|
Yang Y, Wu M, Wegener AJ, Vázquez-Guardado A, Efimov AI, Lie F, Wang T, Ma Y, Banks A, Li Z, Xie Z, Huang Y, Good CH, Kozorovitskiy Y, Rogers JA. Preparation and use of wireless reprogrammable multilateral optogenetic devices for behavioral neuroscience. Nat Protoc 2022; 17:1073-1096. [PMID: 35173306 PMCID: PMC9311268 DOI: 10.1038/s41596-021-00672-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 12/01/2021] [Indexed: 11/08/2022]
Abstract
Wireless battery-free optogenetic devices enable behavioral neuroscience studies in groups of animals with minimal interference to natural behavior. Real-time independent control of optogenetic stimulation through near-field communication dramatically expands the realm of applications of these devices in broad contexts of neuroscience research. Dissemination of these tools with advanced functionalities to the neuroscience community requires protocols for device manufacturing and experimental implementation. This protocol describes detailed procedures for fabrication, encapsulation and implantation of recently developed advanced wireless devices in head- and back-mounted forms. In addition, procedures for standard implementation of experimental systems in mice are provided. This protocol aims to facilitate the application of wireless optogenetic devices in advanced optogenetic experiments involving groups of freely moving rodents and complex environmental designs. The entire protocol lasts ~3-5 weeks.
Collapse
Affiliation(s)
- Yiyuan Yang
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
| | - Mingzheng Wu
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Amy J Wegener
- US Army Research Laboratory, Aberdeen Proving Ground, MD, USA
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA
| | - Abraham Vázquez-Guardado
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
| | - Andrew I Efimov
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | | | - Taoyi Wang
- Department of Precision Instrument, Tsinghua University, Beijing, China
| | - Yuhang Ma
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Anthony Banks
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Neurolux Inc., Evanston, IL, USA
- Feinberg School of Medicine, Northwestern University, Evanston, IL, USA
| | - Zhengwei Li
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
| | - Zhaoqian Xie
- State Key Laboratory of Structural Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
- Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
- Ningbo Institute of Dalian University of Technology, Ningbo, China
| | - Yonggang Huang
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, IL, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
| | - Cameron H Good
- US Army Research Laboratory, Aberdeen Proving Ground, MD, USA.
- US Army Medical Research Institute of Chemical Defense, Aberdeen Proving Ground, MD, USA.
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA.
| | - Yevgenia Kozorovitskiy
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.
- Chemistry of Life Processes Institutes, Northwestern University, Evanston, IL, USA.
| | - John A Rogers
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA.
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA.
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Neurolux Inc., Evanston, IL, USA.
- Feinberg School of Medicine, Northwestern University, Evanston, IL, USA.
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA.
- Department of Chemistry, Northwestern University, Evanston, IL, USA.
- Department of Neurological Surgery, Northwestern University, Evanston, IL, USA.
- Department of Electrical and Computer Engineering, Northwestern University, Evanston, IL, USA.
- Department of Computer Science, Northwestern University, Evanston, IL, USA.
| |
Collapse
|
47
|
Subramanian S, Haroutounian S, Palanca BJA, Lenze EJ. Ketamine as a therapeutic agent for depression and pain: mechanisms and evidence. J Neurol Sci 2022; 434:120152. [PMID: 35092901 DOI: 10.1016/j.jns.2022.120152] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 12/14/2022]
Abstract
Ketamine is an anesthetic drug which is now used to treat chronic pain conditions and psychiatric disorders, especially depression. It is an N-methyl-D-aspartate (NMDA) receptor antagonist with additional effects on α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, opioid receptors, and monoaminergic receptors. This article focuses on ketamine's role in treating depression and pain, two commonly comorbid challenging conditions with potentially shared neurobiologic circuitry. Many clinical trials have utilized intravenous or intranasal ketamine for treating depression and pain. Intravenous ketamine is more bioavailable than intranasal ketamine and both are effective for acute depressive episodes. Intravenous ketamine is advantageous for post-operative analgesia and is associated with a reduction in total opioid requirements. Few studies have treated chronic pain or concurrent depression and pain with ketamine. Larger, randomized control trials are needed to examine the safety and efficacy of intravenous vs. intranasal ketamine, ideal target populations, and optimal dosing to treat both depression and pain.
Collapse
Affiliation(s)
- Subha Subramanian
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| | - Simon Haroutounian
- Department of Anesthesiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Ben Julian A Palanca
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Department of Anesthesiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Eric J Lenze
- Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
48
|
Kang MJY, Hawken E, Vazquez GH. The Mechanisms Behind Rapid Antidepressant Effects of Ketamine: A Systematic Review With a Focus on Molecular Neuroplasticity. Front Psychiatry 2022; 13:860882. [PMID: 35546951 PMCID: PMC9082546 DOI: 10.3389/fpsyt.2022.860882] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/18/2022] [Indexed: 12/25/2022] Open
Abstract
The mechanism of action underlying ketamine's rapid antidepressant effects in patients with depression, both suffering from major depressive disorder (MDD) and bipolar disorder (BD), including treatment resistant depression (TRD), remains unclear. Of the many speculated routes that ketamine may act through, restoring deficits in neuroplasticity may be the most parsimonious mechanism in both human patients and preclinical models of depression. Here, we conducted a literature search using PubMed for any reports of ketamine inducing neuroplasticity relevant to depression, to identify cellular and molecular events, relevant to neuroplasticity, immediately observed with rapid mood improvements in humans or antidepressant-like effects in animals. After screening reports using our inclusion/exclusion criteria, 139 publications with data from cell cultures, animal models, and patients with BD or MDD were included (registered on PROSPERO, ID: CRD42019123346). We found accumulating evidence to support that ketamine induces an increase in molecules involved in modulating neuroplasticity, and that these changes are paired with rapid antidepressant effects. Molecules or complexes of high interest include glutamate, AMPA receptors (AMPAR), mTOR, BDNF/TrkB, VGF, eEF2K, p70S6K, GSK-3, IGF2, Erk, and microRNAs. In summary, these studies suggest a robust relationship between improvements in mood, and ketamine-induced increases in molecular neuroplasticity, particularly regarding intracellular signaling molecules.
Collapse
Affiliation(s)
- Melody J Y Kang
- Center of Neuroscience Studies (CNS), Queen's University, Kingston, ON, Canada
| | - Emily Hawken
- Department of Psychiatry, Queen's University School of Medicine, Kingston, ON, Canada.,Providence Care Hospital, Kingston, ON, Canada
| | - Gustavo Hector Vazquez
- Center of Neuroscience Studies (CNS), Queen's University, Kingston, ON, Canada.,Department of Psychiatry, Queen's University School of Medicine, Kingston, ON, Canada.,Providence Care Hospital, Kingston, ON, Canada
| |
Collapse
|
49
|
Beating Pain with Psychedelics: Matter over Mind? Neurosci Biobehav Rev 2021; 134:104482. [PMID: 34922987 DOI: 10.1016/j.neubiorev.2021.12.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/19/2021] [Accepted: 12/04/2021] [Indexed: 02/08/2023]
Abstract
Basic pain research has shed light on key cellular and molecular mechanisms underlying nociceptive and phenomenological aspects of pain. Despite these advances, [[we still yearn for] the discovery of novel therapeutic strategies to address the unmet needs of about 70% of chronic neuropathic pain patients whose pain fails to respond to opioids as well as to other conventional analgesic agents. Importantly, a substantial body of clinical observations over the past decade cumulatively suggests that the psychedelic class of drugs may possess heuristic value for understanding and treating chronic pain conditions. The present review presents a theoretical framework for hitherto insufficiently understood neuroscience-based mechanisms of psychedelics' potential analgesic effects. To that end, searches of PubMed-indexed journals were performed using the following Medical Subject Headings' terms: pain, analgesia, inflammatory, brain connectivity, ketamine, psilocybin, functional imaging, and dendrites. Recursive sets of scientific and clinical evidence extracted from this literature review were summarized within the following key areas: (1) studies employing psychedelics for alleviation of physical and emotional pain; (2) potential neuro-restorative effects of psychedelics to remediate the impaired connectivity underlying the dissociation between pain-related conscious states/cognitions and the subcortical activity/function leading to the eventual chronicity through immediate and long-term effects on dentritic plasticity; (3) anti-neuroinflammatory and pro-immunomodulatory actions of psychedelics as the may pertain to the role of these factors in the pathogenesis of neuropathic pain; (4) safety, legal, and ethical consideration inherent in psychedelics' pharmacotherapy. In addition to direct beneficial effects in terms of reduction of pain and suffering, psychedelics' inclusion in the analgesic armamentarium will contribute to deeper and more sophisticated insights not only into pain syndromes but also into frequently comorbid psychiatric condition associated with emotional pain, e.g., depressive and anxiety disorders. Further inquiry is clearly warranted into the above areas that have potential to evolve into further elucidate the mechanisms of chronic pain and affective disorders, and lead to the development of innovative, safe, and more efficacious neurobiologically-based therapeutic approaches.
Collapse
|
50
|
Yang Q, Wei T, Yin RT, Wu M, Xu Y, Koo J, Choi YS, Xie Z, Chen SW, Kandela I, Yao S, Deng Y, Avila R, Liu TL, Bai W, Yang Y, Han M, Zhang Q, Haney CR, Benjamin Lee K, Aras K, Wang T, Seo MH, Luan H, Lee SM, Brikha A, Ghoreishi-Haack N, Tran L, Stepien I, Aird F, Waters EA, Yu X, Banks A, Trachiotis GD, Torkelson JM, Huang Y, Kozorovitskiy Y, Efimov IR, Rogers JA. Photocurable bioresorbable adhesives as functional interfaces between flexible bioelectronic devices and soft biological tissues. NATURE MATERIALS 2021; 20:1559-1570. [PMID: 34326506 PMCID: PMC8551016 DOI: 10.1038/s41563-021-01051-x] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 06/11/2021] [Indexed: 05/07/2023]
Abstract
Flexible electronic/optoelectronic systems that can intimately integrate onto the surfaces of vital organ systems have the potential to offer revolutionary diagnostic and therapeutic capabilities relevant to a wide spectrum of diseases and disorders. The critical interfaces between such technologies and living tissues must provide soft mechanical coupling and efficient optical/electrical/chemical exchange. Here, we introduce a functional adhesive bioelectronic-tissue interface material, in the forms of mechanically compliant, electrically conductive, and optically transparent encapsulating coatings, interfacial layers or supporting matrices. These materials strongly bond both to the surfaces of the devices and to those of different internal organs, with stable adhesion for several days to months, in chemistries that can be tailored to bioresorb at controlled rates. Experimental demonstrations in live animal models include device applications that range from battery-free optoelectronic systems for deep-brain optogenetics and subdermal phototherapy to wireless millimetre-scale pacemakers and flexible multielectrode epicardial arrays. These advances have immediate applicability across nearly all types of bioelectronic/optoelectronic system currently used in animal model studies, and they also have the potential for future treatment of life-threatening diseases and disorders in humans.
Collapse
Affiliation(s)
- Quansan Yang
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
| | - Tong Wei
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, USA
| | - Rose T Yin
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - Mingzheng Wu
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Yameng Xu
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
- The Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Jahyun Koo
- School of Biomedical Engineering, Korea University, Seoul, Republic of Korea
| | - Yeon Sik Choi
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
| | - Zhaoqian Xie
- State Key Laboratory of Structural Analysis for Industrial Equipment, Dalian University of Technology, Dalian, China
- Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
- Ningbo Institute of Dalian University of Technology, Ningbo, China
| | - Sheena W Chen
- Department of Surgery, The George Washington University, Washington, DC, USA
| | - Irawati Kandela
- Developmental Therapeutics Core, Northwestern University, Evanston, IL, USA
- Chemistry Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Shenglian Yao
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing, China
| | - Yujun Deng
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
- State Key Laboratory of Mechanical System and Vibration, Shanghai Jiao Tong University, Shanghai, China
| | - Raudel Avila
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
| | - Tzu-Li Liu
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
| | - Wubin Bai
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yiyuan Yang
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
| | - Mengdi Han
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
| | - Qihui Zhang
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
| | - Chad R Haney
- Chemistry Life Processes Institute, Northwestern University, Evanston, IL, USA
- Center for Advanced Molecular Imaging, Northwestern University, Evanston, IL, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - K Benjamin Lee
- Department of Surgery, The George Washington University, Washington, DC, USA
| | - Kedar Aras
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - Tong Wang
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, USA
| | - Min-Ho Seo
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
- School of Biomedical Convergence Engineering, College of Information & Biomedical Engineering, Pusan National University, Pusan, Republic of Korea
| | - Haiwen Luan
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
| | - Seung Min Lee
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
| | - Anlil Brikha
- Chemistry Life Processes Institute, Northwestern University, Evanston, IL, USA
- Center for Advanced Molecular Imaging, Northwestern University, Evanston, IL, USA
| | | | - Lori Tran
- Developmental Therapeutics Core, Northwestern University, Evanston, IL, USA
- Chemistry Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Iwona Stepien
- Developmental Therapeutics Core, Northwestern University, Evanston, IL, USA
- Chemistry Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Fraser Aird
- Chemistry Life Processes Institute, Northwestern University, Evanston, IL, USA
| | - Emily A Waters
- Chemistry Life Processes Institute, Northwestern University, Evanston, IL, USA
- Center for Advanced Molecular Imaging, Northwestern University, Evanston, IL, USA
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Xinge Yu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Anthony Banks
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA
| | - Gregory D Trachiotis
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
- DC Veterans Affairs Medical Center, The George Washington University, Washington, DC, USA
| | - John M Torkelson
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
| | - Yonggang Huang
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA
- Departments of Civil and Environmental Engineering, Northwestern University, Evanston, IL, USA
| | - Yevgenia Kozorovitskiy
- Department of Neurobiology, Northwestern University, Evanston, IL, USA.
- Chemistry Life Processes Institute, Northwestern University, Evanston, IL, USA.
| | - Igor R Efimov
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA.
| | - John A Rogers
- Center for Bio-Integrated Electronics, Northwestern University, Evanston, IL, USA.
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, IL, USA.
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, USA.
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, USA.
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|