1
|
Sun Y, Wang X, Chen Y, Luan Z, Hao R. The impact of exogenous Oxytocin on visual cortex plasticity across different stages of visual development. Sci Rep 2025; 15:12137. [PMID: 40204929 PMCID: PMC11982226 DOI: 10.1038/s41598-025-96573-8] [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: 12/01/2024] [Accepted: 03/31/2025] [Indexed: 04/11/2025] Open
Abstract
The plasticity of ocular dominance is most prominent during the critical period of visual development, influenced by the balance of excitatory and inhibitory synaptic transmission in the visual cortex. Astrocytes play a crucial role in regulating synaptic plasticity through phagocytosis of synapses. However, the ability of astrocytes to modulate synaptic plasticity after the critical period remains unclear. Oxytocin (OT), a neuropeptide involved in neural circuit formation, has shown potential in enhancing synaptic plasticity. This study explores the role of OT in restoring visual cortical plasticity during and after the critical period of visual development. We performed monocular deprivation (MD) on mice during the critical period and extended the deprivation until adulthood. Visual cortical plasticity was evaluated using pattern visual evoked potentials (PVEPs), immunofluorescence staining, and western blotting. Excitatory synaptic markers (VGLUT1, PSD- 95) and inhibitory synaptic markers (VGAT, Gephyrin) were analyzed. The effects of OT administration, alone or combined with reverse occlusion (RO), on ocular dominance plasticity and astrocyte activity were assessed. During the critical period, MD induced a significant ocular dominance shift with reduced cortical response from the deprived eye, primarily through decreased excitatory synaptic markers (VGLUT1: P < 0.05; PSD- 95: P < 0.05). OT administration further enhanced this shift by reducing GFAP expression and decreasing astrocytic phagocytosis of excitatory synapses. After the critical period, prolonged MD reduced excitatory synaptic marker expression in the visual cortex (P < 0.05), and RO alone did not restore cortical plasticity. However, the combination of OT and RO increased excitatory synaptic marker expression (VGLUT1: P < 0.05; PSD- 95: P < 0.05 and restored ocular dominance plasticity. Our findings demonstrate that OT can modulate astrocyte activity and enhance excitatory synaptic plasticity, facilitating the recovery of visual cortical plasticity both during and after the critical period. These results highlight the potential of OT as a therapeutic intervention for visual impairments caused by disrupted sensory experiences during development.
Collapse
Affiliation(s)
- Yifan Sun
- Clinical College of Ophthalmology, Tianjin Medical University, 300020, Tianjin, PR China
- Tianjin Key Lab of Ophthalmology and Vision Science, Tianjin Eye Hospital, Tianjin Eye Institute, 300020, Tianjin, PR China
| | - Xiao Wang
- Clinical College of Ophthalmology, Tianjin Medical University, 300020, Tianjin, PR China
- Tianjin Key Lab of Ophthalmology and Vision Science, Tianjin Eye Hospital, Tianjin Eye Institute, 300020, Tianjin, PR China
| | - Yamin Chen
- Clinical College of Ophthalmology, Tianjin Medical University, 300020, Tianjin, PR China
- Tianjin Key Lab of Ophthalmology and Vision Science, Tianjin Eye Hospital, Tianjin Eye Institute, 300020, Tianjin, PR China
| | - Zichen Luan
- Clinical College of Ophthalmology, Tianjin Medical University, 300020, Tianjin, PR China
- Tianjin Key Lab of Ophthalmology and Vision Science, Tianjin Eye Hospital, Tianjin Eye Institute, 300020, Tianjin, PR China
| | - Rui Hao
- Clinical College of Ophthalmology, Tianjin Medical University, 300020, Tianjin, PR China.
- Tianjin Key Lab of Ophthalmology and Vision Science, Tianjin Eye Hospital, Tianjin Eye Institute, 300020, Tianjin, PR China.
- Nankai University Affiliated Eye Hospital, Nankai University, 300020, Tianjin, PR China.
| |
Collapse
|
2
|
Ding S, Liu Y, Tao H, Zhao Y, Zeng H, Han Y, Wang S, Chen Z, Tang Y, Guo W. Chronic intranasal oxytocin alleviates cognitive impairment and reverses oxytocin signaling upregulation in MK801-induced mice. Psychoneuroendocrinology 2024; 168:107138. [PMID: 39068687 DOI: 10.1016/j.psyneuen.2024.107138] [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: 01/03/2024] [Revised: 07/03/2024] [Accepted: 07/13/2024] [Indexed: 07/30/2024]
Abstract
OBJECTIVE Cognitive impairment, especially impaired social cognition, is largely responsible for the deterioration of the social life of patients with schizophrenia (SZ). Oxytocin (OT) is a neuropeptide that offers promising therapy for SZ. This study aimed to explore whether OT could affect dizocilpine (MK801)-induced cognitive impairment and to investigate the effect of exogenous OT on the endogenous OT system in the hippocampus. METHODS The SZ mouse model was established by repeated administration of dizocilpine [MK801, 0.6 mg/kg, intraperitoneal (i.p.)], and then OT (6-60 μg/kg, intranasal) or risperidone (0.3 mg/kg, i.p.) was administered to explore the effect of OT on cognitive impairment. RESULTS OT at a dose of 6 μg/kg alleviated MK801-induced hyperactivity, sociability impairment, and spatial memory impairment. OT at a dose of 20 or 60 μg/kg attenuated the hyperactivity and social novelty impairment. In MK801-injected mice, the compensatory upregulation of OT mRNA in the hippocampus was reversed by three OT doses, whereas 60 μg/kg OT reversed the compensatory upregulation of CD38 protein expression. CONCLUSION OT alleviated cognitive impairment in the SZ mouse model to varying degrees, reversing the compensatory upregulation of OT signaling in the hippocampus.
Collapse
Affiliation(s)
- Shan Ding
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yong Liu
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Huai Tao
- Department of Biochemistry and Molecular Biology, Hunan University of Chinese Medicine, Changsha, China
| | - Yuxu Zhao
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Hongtao Zeng
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yiding Han
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shichen Wang
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiheng Chen
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yamei Tang
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, China.
| | - Wenbin Guo
- Department of Psychiatry, National Clinical Research Center for Mental Disorders, and National Center for Mental Disorders, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
3
|
Winter J, Meyer M, Berger I, Royer M, Bianchi M, Kuffner K, Peters S, Stang S, Langgartner D, Hartmann F, Schmidtner AK, Reber SO, Bosch OJ, Bludau A, Slattery DA, van den Burg EH, Jurek B, Neumann ID. Chronic oxytocin-driven alternative splicing of Crfr2α induces anxiety. Mol Psychiatry 2023; 28:4742-4755. [PMID: 34035479 PMCID: PMC10914602 DOI: 10.1038/s41380-021-01141-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/25/2021] [Accepted: 04/20/2021] [Indexed: 12/26/2022]
Abstract
The neuropeptide oxytocin (OXT) has generated considerable interest as potential treatment for psychiatric disorders, including anxiety and autism spectrum disorders. However, the behavioral and molecular consequences associated with chronic OXT treatment and chronic receptor (OXTR) activation have scarcely been studied, despite the potential therapeutic long-term use of intranasal OXT. Here, we reveal that chronic OXT treatment over two weeks increased anxiety-like behavior in rats, with higher sensitivity in females, contrasting the well-known anxiolytic effect of acute OXT. The increase in anxiety was transient and waned 5 days after the infusion has ended. The behavioral effects of chronic OXT were paralleled by activation of an intracellular signaling pathway, which ultimately led to alternative splicing of hypothalamic corticotropin-releasing factor receptor 2α (Crfr2α), an important modulator of anxiety. In detail, chronic OXT shifted the splicing ratio from the anxiolytic membrane-bound (mCRFR2α) form of CRFR2α towards the soluble CRFR2α (sCRFR2α) form. Experimental induction of alternative splicing mimicked the anxiogenic effects of chronic OXT, while sCRFR2α-knock down reduced anxiety-related behavior of male rats. Furthermore, chronic OXT treatment triggered the release of sCRFR2α into the cerebrospinal fluid with sCRFR2α levels positively correlating with anxiety-like behavior. In summary, we revealed that the shifted splicing ratio towards expression of the anxiogenic sCRFR2α underlies the adverse effects of chronic OXT treatment on anxiety.
Collapse
Affiliation(s)
- Julia Winter
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Magdalena Meyer
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Ilona Berger
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Melanie Royer
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Marta Bianchi
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Kerstin Kuffner
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Sebastian Peters
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Simone Stang
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Dominik Langgartner
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, University of Ulm, Ulm, Germany
| | - Finn Hartmann
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Anna K Schmidtner
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Stefan O Reber
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, University of Ulm, Ulm, Germany
| | - Oliver J Bosch
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Anna Bludau
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - David A Slattery
- Laboratory of Translational Psychiatry, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University of Frankfurt, Frankfurt am Main, Germany
| | - Erwin H van den Burg
- Center for Psychiatric Neurosciences, University Hospital Lausanne, Lausanne, Switzerland
| | - Benjamin Jurek
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany
| | - Inga D Neumann
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
4
|
Gozzi A, Zerbi V. Modeling Brain Dysconnectivity in Rodents. Biol Psychiatry 2023; 93:419-429. [PMID: 36517282 DOI: 10.1016/j.biopsych.2022.09.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/19/2022] [Accepted: 09/10/2022] [Indexed: 02/04/2023]
Abstract
Altered or atypical functional connectivity as measured with functional magnetic resonance imaging (fMRI) is a hallmark feature of brain connectopathy in psychiatric, developmental, and neurological disorders. However, the biological underpinnings and etiopathological significance of this phenomenon remain unclear. The recent development of MRI-based techniques for mapping brain function in rodents provides a powerful platform to uncover the determinants of functional (dys)connectivity, whether they are genetic mutations, environmental risk factors, or specific cellular and circuit dysfunctions. Here, we summarize the recent contribution of rodent fMRI toward a deeper understanding of network dysconnectivity in developmental and psychiatric disorders. We highlight substantial correspondences in the spatiotemporal organization of rodent and human fMRI networks, supporting the translational relevance of this approach. We then show how this research platform might help us comprehend the importance of connectional heterogeneity in complex brain disorders and causally relate multiscale pathogenic contributors to functional dysconnectivity patterns. Finally, we explore how perturbational techniques can be used to dissect the fundamental aspects of fMRI coupling and reveal the causal contribution of neuromodulatory systems to macroscale network activity, as well as its altered dynamics in brain diseases. These examples outline how rodent functional imaging is poised to advance our understanding of the bases and determinants of human functional dysconnectivity.
Collapse
Affiliation(s)
- Alessandro Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems, Rovereto, Italy.
| | - Valerio Zerbi
- Neuro-X Institute, School of Engineering, École polytechnique fédérale de Lausanne, Lausanne, Switzerland; CIBM Center for Biomedical Imaging, Lausanne, Switzerland.
| |
Collapse
|
5
|
Finton CJ, Ophir AG. Developmental exposure to intranasal vasopressin impacts adult prairie vole spatial memory. Psychoneuroendocrinology 2022; 141:105750. [PMID: 35397260 PMCID: PMC9149121 DOI: 10.1016/j.psyneuen.2022.105750] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/22/2022] [Accepted: 03/22/2022] [Indexed: 11/18/2022]
Abstract
Spatial memory is critical for many tasks necessary for survival (i.e., locating mates and food resources). The two mammalian nonapeptides arginine vasopressin (AVP) and oxytocin (OT) are mechanistically important in modulating memory ability, albeit in contrasting ways. In general, AVP facilitates memory consolidation and retrieval while OT is an amnesic. Although AVP and OT are known to have these memory effects, past work has focused on their impact in social memory with little research on their effects on spatial memory. In this experiment, we tested the impact of AVP and OT on spatial memory as determined by performance in the Morris water maze (MWM). We administered doses of AVP, OT, or saline (a control) intranasally to male prairie voles (Microtus ochrogaster), a species whose spatial memory is hypothesized to impact their mating tactics. We also investigated if acute doses (given immediately prior to the memory trial in the MWM) and chronic doses (given daily during adolescence) had differing impacts on spatial cognition. We found that chronic intranasal administration of AVP during post-wean development improved spatial memory performance. In contrast, both chronic and acute administration of OT and acute administration of AVP had no impact on spatial memory. These results together suggest that 1) chronic exposure to AVP has organizational effects on spatial memory in the prairie vole, and 2) acute administration of nonapeptides does not impact the retrieval of spatial memories.
Collapse
Affiliation(s)
- Caitlyn J Finton
- Department of Psychology, Cornell University, Ithaca, NY 14850, USA
| | - Alexander G Ophir
- Department of Psychology, Cornell University, Ithaca, NY 14850, USA.
| |
Collapse
|
6
|
Lindenmaier Z, Ellegood J, Stuive M, Easson K, Yee Y, Fernandes D, Foster J, Anagnostou E, Lerch JP. Examining the effect of chronic intranasal oxytocin administration on the neuroanatomy and behavior of three autism-related mouse models. Neuroimage 2022; 257:119243. [PMID: 35508216 DOI: 10.1016/j.neuroimage.2022.119243] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/14/2022] [Accepted: 04/21/2022] [Indexed: 11/28/2022] Open
Abstract
Although initially showing great potential, oxytocin treatment has encountered a translational hurdle in its promise of treating the social deficits of autism. Some debate surrounds the ability of oxytocin to successfully enter the brain, and therefore modify neuroanatomy. Moreover, given the heterogeneous nature of autism, treatment will only amerliorate symptoms in a subset of patients. Therefore, to determine whether oxytocin changes brain circuitry, and whether it does so variably, depending on genotype, we implemented a large randomized, blinded, placebo-controlled, preclinical study on chronic intranasal oxytocin treatment in three different mouse models related to autism with a focus on using neuroanatomical phenotypes to assess and subset treatment response. Intranasal oxytocin (0.6IU) was administered daily, for 28 days, starting at 5 weeks of age to the 16p11.2 deletion, Shank3 (exon 4-9) knockout, and Fmr1 knockout mouse models. Given the sensitivity of structural magnetic resonance imaging (MRI) to the neurological effects of interventions like drugs, along with many other advantages, the mice underwent in vivo longitudinal and high-resolution ex vivo imaging with MRI. The scans included three in vivo T1weighted, 90um isotropic resolution scans and a T2-weighted, 3D fast spin echo with 40um isotropic resolution ex vivo scan to assess the changes in neuroanatomy using established automated image registration and deformation based morphometry approaches in response to oxytocin treatment. The behavior of the mice was assessed in multiple domains, including social behaviours and repetitive behaviours, among others. Treatment effect on the neuroanatomy did not reach significance, although the pattern of trending effects was promising. No significant effect of treatment was found on social behavior in any of the strains, although a significant effect of treatment was found in the Fmr1 mouse, with treatment normalizing a grooming deficit. No other treatment effect on behavior was observed that survived multiple comparisons correction. Overall, chronic treatment with oxytocin had limited effects on the three mouse models related to autism, and no promising pattern of response susceptibility emerged.
Collapse
Affiliation(s)
- Zsuzsa Lindenmaier
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| | - Jacob Ellegood
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Monique Stuive
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kaitlyn Easson
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Yohan Yee
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Darren Fernandes
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Jane Foster
- Department of Psychiatry and Behavioral Neurosciences, McMaster University, St.Joseph's Healthcare, Hamilton, Ontario, Canada
| | - Evdokia Anagnostou
- Autism Research Center, Holland Bloorview Kids Rehabilitation Hospital, Toronto, Canada
| | - Jason P Lerch
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Wellcome Centre for Integrative NeuroImaging, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
7
|
Martins D, Brodmann K, Veronese M, Dipasquale O, Mazibuko N, Schuschnig U, Zelaya F, Fotopoulou A, Paloyelis Y. "Less is more": a dose-response account of intranasal oxytocin pharmacodynamics in the human brain. Prog Neurobiol 2022; 211:102239. [PMID: 35122880 DOI: 10.1016/j.pneurobio.2022.102239] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/23/2022] [Accepted: 01/31/2022] [Indexed: 12/27/2022]
Abstract
Intranasal oxytocin is attracting attention as a potential treatment for several brain disorders due to promising preclinical results. However, translating findings to humans has been hampered by remaining uncertainties about its pharmacodynamics and the methods used to probe its effects in the human brain. Using a dose-response design (9, 18 and 36 IU), we demonstrate that intranasal oxytocin-induced changes in local regional cerebral blood flow (rCBF) in the amygdala at rest, and in the covariance between rCBF in the amygdala and other key hubs of the brain oxytocin system, follow a dose-response curve with maximal effects for lower doses. Yet, the effects on local rCBF might vary by amygdala subdivision, highlighting the need to qualify dose-response curves within subregion. We further link physiological changes with the density of the oxytocin receptor gene mRNA across brain regions, strengthening our confidence in intranasal oxytocin as a valid approach to engage central targets. Finally, we demonstrate that intranasal oxytocin does not disrupt cerebrovascular reactivity, which corroborates the validity of haemodynamic neuroimaging to probe the effects of intranasal oxytocin in the human brain. DATA AVAILABILITY: Participants did not consent for open sharing of the data. Therefore, data can only be accessed from the corresponding author upon reasonable request.
Collapse
Affiliation(s)
- Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Katja Brodmann
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Ottavia Dipasquale
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Ndaba Mazibuko
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | | | - Fernando Zelaya
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Aikaterini Fotopoulou
- Department of Clinical, Educational and Health Psychology, University College London, London, UK
| | - Yannis Paloyelis
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK.
| |
Collapse
|
8
|
Yamasue H, Kojima M, Kuwabara H, Kuroda M, Matsumoto K, Kanai C, Inada N, Owada K, Ochi K, Ono N, Benner S, Wakuda T, Kameno Y, Inoue J, Harada T, Tsuchiya K, Umemura K, Yamauchi A, Ogawa N, Kushima I, Ozaki N, Suyama S, Saito T, Uemura Y, Hamada J, Kano Y, Honda N, Kikuchi S, Seto M, Tomita H, Miyoshi N, Matsumoto M, Kawaguchi Y, Kanai K, Ikeda M, Nakamura I, Isomura S, Hirano Y, Onitsuka T, Kosaka H, Okada T. Effect of a novel nasal oxytocin spray with enhanced bioavailability on autism: a randomized trial. Brain 2022; 145:490-499. [DOI: 10.1093/brain/awab291] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/14/2021] [Accepted: 07/14/2021] [Indexed: 12/13/2022] Open
Abstract
Abstract
Although intranasal oxytocin is expected to be a novel therapy for the core symptoms of autism spectrum disorder, which has currently no approved medication, the efficacy of repeated administrations was inconsistent, suggesting that the optimal dose for a single administration of oxytocin is not optimal for repeated administration.
The current double-blind, placebo-controlled, multicentre, crossover trial (ClinicalTrials.gov Identifier: NCT03466671) was aimed to test the effect of TTA-121, a new formulation of intranasal oxytocin spray with an enhanced bioavailability (3.6 times higher than Syntocinon® spray, as assessed by area under the concentration–time curve in rabbit brains), which enabled us to test a wide range of multiple doses, on autism spectrum disorder core symptoms and to determine the dose–response relationship. Four-week administrations of TTA-121, at low dose once per day (3 U/day), low dose twice per day (6 U/day), high dose once per day (10 U/day), or high dose twice per day (20 U/day), and 4-week placebo were administered in a crossover manner. The primary outcome was the mean difference in the reciprocity score (range: 0–14, higher values represent worse outcomes) on the Autism Diagnostic Observation Schedule between the baseline and end point of each administration period. This trial with two administration periods and eight groups was conducted at seven university hospitals in Japan, enrolling adult males with high-functioning autism spectrum disorder. Enrolment began from June 2018 and ended December 2019. Follow-up ended March 2020.
Of 109 males with high-functioning autism spectrum disorder who were randomized, 103 completed the trial. The smallest P-value, judged as the dose–response relationship, was the contrast with the peak at TTA-121 6 U/day, with inverted U-shape for both the full analysis set (P = 0.182) and per protocol set (P = 0.073). The Autism Diagnostic Observation Schedule reciprocity score, the primary outcome, was reduced in the TTA-121 6 U/day administration period compared with the placebo (full analysis set: P = 0.118, mean difference = −0.5; 95% CI: −1.1 to 0.1; per protocol set: P = 0.012, mean difference = −0.8; 95% CI: −1.3 to −0.2). The per protocol set was the analysis target population, consisting of all full analysis set participants except those who deviated from the protocol. Most dropouts from the full analysis set to the per protocol set occurred because of poor adherence to the test drug (9 of 12 in the first period and 8 of 15 in the second period). None of the secondary clinical and behavioural outcomes were significantly improved with the TTA-121 compared with the placebo in the full analysis set.
A novel intranasal spray of oxytocin with enhanced bioavailability enabled us to test a wide range of multiple doses, revealing an inverted U-shape dose–response curve, with the peak at a dose that was lower than expected from previous studies. The efficacy of TTA-121 shown in the current exploratory study should be verified in a future large-scale, parallel-group trial.
Collapse
Affiliation(s)
- Hidenori Yamasue
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu City 431-3192, Japan
- Department of Child Development, United Graduate School of Child Development at Hamamatsu, 1-20-1 Handayama, Higashiku, Hamamatsu 431-3192, Japan
| | - Masaki Kojima
- Department of Child Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Hitoshi Kuwabara
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu City 431-3192, Japan
- Department of Child Development, United Graduate School of Child Development at Hamamatsu, 1-20-1 Handayama, Higashiku, Hamamatsu 431-3192, Japan
| | - Miho Kuroda
- Department of Psychology, Faculty of Liberal Arts, Teikyo University, Tokyo, Japan
| | - Kaori Matsumoto
- Graduate School of Psychology, Kanazawa Institute of Technology, 7-1 Ohgigaoka, Nonoichi 921-8054, Japan
| | - Chieko Kanai
- Child Development and Education, Faculty of Humanities, Wayo Women’s University, Konodai 2-3-1, Ichikawa, Chiba 272-0827, Japan
| | - Naoko Inada
- Department of Psychology, Faculty of Liberal Arts, Teikyo University, Tokyo, Japan
| | - Keiho Owada
- Department of Pediatrics, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Keiko Ochi
- School of Media Science, Tokyo University of Technology, Hachioji, Japan
| | - Nobutaka Ono
- Department of Computer Science, Graduate School of Systems Design, Tokyo Metropolitan University, Hino, Japan
| | - Seico Benner
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu City 431-3192, Japan
| | - Tomoyasu Wakuda
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu City 431-3192, Japan
| | - Yosuke Kameno
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu City 431-3192, Japan
| | - Jun Inoue
- Department of Child and Adolescent Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu City 431-3192, Japan
| | - Taeko Harada
- Department of Child Development, United Graduate School of Child Development at Hamamatsu, 1-20-1 Handayama, Higashiku, Hamamatsu 431-3192, Japan
| | - Kenji Tsuchiya
- Department of Child Development, United Graduate School of Child Development at Hamamatsu, 1-20-1 Handayama, Higashiku, Hamamatsu 431-3192, Japan
| | - Kazuo Umemura
- Department of Pharmacology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu City 431-3192, Japan
| | - Aya Yamauchi
- Department of Medical Technique, Nagoya University Hospital, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8560, Japan
| | - Nanayo Ogawa
- Department of Psychiatry, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | - Itaru Kushima
- Department of Psychiatry, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | - Satoshi Suyama
- Department of Child and Adolescent Psychiatry, Hokkaido University Hospital, Sapporo, Japan
| | - Takuya Saito
- Department of Child and Adolescent Psychiatry, Hokkaido University Hospital, Sapporo, Japan
| | - Yukari Uemura
- Biostatistics Section, Department of Data Science, Center for Clinical Science, National Center for Global Health and Medicine, Shinjyu-ku, Tokyo 162-8655, Japan
| | - Junko Hamada
- Department of Child Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yukiko Kano
- Department of Child Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Nami Honda
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Saya Kikuchi
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Moe Seto
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Hiroaki Tomita
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Noriko Miyoshi
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Megumi Matsumoto
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuko Kawaguchi
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Koji Kanai
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Itta Nakamura
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shuichi Isomura
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoji Hirano
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshiaki Onitsuka
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hirotaka Kosaka
- Department of Neuropsychiatry, Faculty of Medical Sciences, University of Fukui, Eiheiji, Fukui 910-1193, Japan
| | - Takashi Okada
- Department of Psychiatry, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| |
Collapse
|
9
|
Ghazy AA, Soliman OA, Elbahnasi AI, Alawy AY, Mansour AM, Gowayed MA. Role of Oxytocin in Different Neuropsychiatric, Neurodegenerative, and Neurodevelopmental Disorders. Rev Physiol Biochem Pharmacol 2022; 186:95-134. [PMID: 36416982 DOI: 10.1007/112_2022_72] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Oxytocin has recently gained significant attention because of its role in the pathophysiology and management of dominant neuropsychiatric disorders. Oxytocin, a peptide hormone synthesized in the hypothalamus, is released into different brain regions, acting as a neurotransmitter. Receptors for oxytocin are present in many areas of the brain, including the hypothalamus, amygdala, and nucleus accumbens, which have been involved in the pathophysiology of depression, anxiety, schizophrenia, autism, Alzheimer's disease, Parkinson's disease, and attention deficit hyperactivity disorder. Animal studies have spotlighted the role of oxytocin in social, behavioral, pair bonding, and mother-infant bonding. Furthermore, oxytocin protects fetal neurons against injury during childbirth and affects various behaviors, assuming its possible neuroprotective characteristics. In this review, we discuss some of the concepts and mechanisms related to the role of oxytocin in the pathophysiology and management of some neuropsychiatric, neurodegenerative, and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Aya A Ghazy
- Department of Clinical Pharmacy, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Omar A Soliman
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Aya I Elbahnasi
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Aya Y Alawy
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Amira Ma Mansour
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Mennatallah A Gowayed
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt.
| |
Collapse
|
10
|
Pagani M, Barsotti N, Bertero A, Trakoshis S, Ulysse L, Locarno A, Miseviciute I, De Felice A, Canella C, Supekar K, Galbusera A, Menon V, Tonini R, Deco G, Lombardo MV, Pasqualetti M, Gozzi A. mTOR-related synaptic pathology causes autism spectrum disorder-associated functional hyperconnectivity. Nat Commun 2021; 12:6084. [PMID: 34667149 PMCID: PMC8526836 DOI: 10.1038/s41467-021-26131-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 09/17/2021] [Indexed: 11/24/2022] Open
Abstract
Postmortem studies have revealed increased density of excitatory synapses in the brains of individuals with autism spectrum disorder (ASD), with a putative link to aberrant mTOR-dependent synaptic pruning. ASD is also characterized by atypical macroscale functional connectivity as measured with resting-state fMRI (rsfMRI). These observations raise the question of whether excess of synapses causes aberrant functional connectivity in ASD. Using rsfMRI, electrophysiology and in silico modelling in Tsc2 haploinsufficient mice, we show that mTOR-dependent increased spine density is associated with ASD -like stereotypies and cortico-striatal hyperconnectivity. These deficits are completely rescued by pharmacological inhibition of mTOR. Notably, we further demonstrate that children with idiopathic ASD exhibit analogous cortical-striatal hyperconnectivity, and document that this connectivity fingerprint is enriched for ASD-dysregulated genes interacting with mTOR or Tsc2. Finally, we show that the identified transcriptomic signature is predominantly expressed in a subset of children with autism, thereby defining a segregable autism subtype. Our findings causally link mTOR-related synaptic pathology to large-scale network aberrations, revealing a unifying multi-scale framework that mechanistically reconciles developmental synaptopathy and functional hyperconnectivity in autism.
Collapse
Affiliation(s)
- Marco Pagani
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
- Autism Center, Child Mind Institute, New York, NY, USA
| | - Noemi Barsotti
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy
| | - Alice Bertero
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy
| | - Stavros Trakoshis
- Department of Psychology, University of Cyprus, Nicosia, Cyprus
- Laboratory for Autism and Neurodevelopmental Disorders, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
| | - Laura Ulysse
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Barcelona, Spain
| | - Andrea Locarno
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | - Ieva Miseviciute
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | - Alessia De Felice
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
| | - Carola Canella
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
| | | | - Alberto Galbusera
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
| | | | - Raffaella Tonini
- Neuromodulation of Cortical and Subcortical Circuits Laboratory, Istituto Italiano di Tecnologia, Genova, Italy
| | - Gustavo Deco
- Center for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Barcelona, Spain
- Institució Catalana de la Recerca i Estudis Avançats (ICREA), Universitat Pompeu Fabra, Barcelona, Spain
| | - Michael V Lombardo
- Laboratory for Autism and Neurodevelopmental Disorders, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
- Autism Research Centre, University of Cambridge, Cambridge, UK
| | - Massimo Pasqualetti
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy
- Department of Biology, Unit of Cell and Developmental Biology, University of Pisa, Pisa, Italy
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Istituto Italiano di Tecnologia, Center for Neuroscience and Cognitive Systems @ University of Trento, Rovereto, Italy.
| |
Collapse
|
11
|
Fastman J, Foss-Feig J, Frank Y, Halpern D, Harony-Nicolas H, Layton C, Sandin S, Siper P, Tang L, Trelles P, Zweifach J, Buxbaum JD, Kolevzon A. A randomized controlled trial of intranasal oxytocin in Phelan-McDermid syndrome. Mol Autism 2021; 12:62. [PMID: 34593045 PMCID: PMC8482590 DOI: 10.1186/s13229-021-00459-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/19/2021] [Indexed: 12/20/2022] Open
Abstract
Background Phelan-McDermid syndrome (PMS) is a rare neurodevelopmental disorder caused by haploinsufficiency of the SHANK3 gene and characterized by global developmental delays, deficits in speech and motor function, and autism spectrum disorder (ASD). Monogenic causes of ASD such as PMS are well suited to investigations with novel therapeutics, as interventions can be targeted based on established genetic etiology. While preclinical studies have demonstrated that the neuropeptide oxytocin can reverse electrophysiological, attentional, and social recognition memory deficits in Shank3-deficient rats, there have been no trials in individuals with PMS. The purpose of this study is to assess the efficacy and safety of intranasal oxytocin as a treatment for the core symptoms of ASD in a cohort of children with PMS. Methods Eighteen children aged 5–17 with PMS were enrolled. Participants were randomized to receive intranasal oxytocin or placebo (intranasal saline) and underwent treatment during a 12-week double-blind, parallel group phase, followed by a 12-week open-label extension phase during which all participants received oxytocin. Efficacy was assessed using the primary outcome of the Aberrant Behavior Checklist-Social Withdrawal (ABC-SW) subscale as well as a number of secondary outcome measures related to the core symptoms of ASD. Safety was monitored throughout the study period. Results There was no statistically significant improvement with oxytocin as compared to placebo on the ABC-SW (Mann–Whitney U = 50, p = 0.055), or on any secondary outcome measures, during either the double-blind or open-label phases. Oxytocin was generally well tolerated, and there were no serious adverse events.
Limitations The small sample size, potential challenges with drug administration, and expectancy bias due to relying on parent reported outcome measures may all contribute to limitations in interpreting results. Conclusion Our results suggest that intranasal oxytocin is not efficacious in improving the core symptoms of ASD in children with PMS. Trial registration NCT02710084. Supplementary Information The online version contains supplementary material available at 10.1186/s13229-021-00459-1.
Collapse
Affiliation(s)
- J Fastman
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J Foss-Feig
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Y Frank
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - D Halpern
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - H Harony-Nicolas
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - C Layton
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - S Sandin
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - P Siper
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - L Tang
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - P Trelles
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J Zweifach
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J D Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - A Kolevzon
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1230, New York, NY, 10029, USA. .,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
12
|
Chronic oxytocin administration in older men modulates functional connectivity during animacy perception. AGING BRAIN 2021; 1:100023. [PMID: 36911518 PMCID: PMC9997159 DOI: 10.1016/j.nbas.2021.100023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 01/21/2023] Open
Abstract
While aging is associated with social-cognitive change and oxytocin plays a crucial role in social cognition, oxytocin's effects on the social brain in older age remain understudied. To date, no study has examined the effects of chronic intranasal oxytocin administration on brain mechanisms underlying animacy perception in older adults. Using a placebo-controlled, randomized, double-blinded design in generally healthy older men (mean age (SD) = 69(6); n = 17 oxytocin; n = 14 placebo), this study determined the effects of a four-week intranasal oxytocin administration (24 international units/twice a day) on functional MRI (fMRI) during the Heider-Simmel task. This passive-viewing animacy perception paradigm contains video-clips of simple shapes suggesting social interactions (SOCIAL condition) or exhibiting random trajectories (RANDOM condition). While there were no oxytocin-specific effects on brain fMRI activation during the SOCIAL compared to the RANDOM condition, pre-to-post intervention change in the SOCIAL-RANDOM difference in functional connectivity (FC) was higher in the oxytocin compared to the placebo group in a network covering occipital, temporal, and parietal areas, and the superior temporal sulcus, a key structure in animacy perception. These findings suggest oxytocin modulation of circuits involved in action observation and social perception. Follow-up analyses on this network's connections suggested a pre-to-post intervention decrease in the SOCIAL-RANDOM difference in FC among the placebo group, possibly reflecting habituation to repeated exposure to social cues. Chronic oxytocin appeared to counter this process by decreasing FC during the RANDOM and increasing it during the SOCIAL condition. This study advances knowledge about oxytocin intervention mechanisms in the social brain of older adults.
Collapse
|
13
|
Brain oxytocin: how puzzle stones from animal studies translate into psychiatry. Mol Psychiatry 2021; 26:265-279. [PMID: 32514104 PMCID: PMC7278240 DOI: 10.1038/s41380-020-0802-9] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/14/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023]
Abstract
The neuropeptide oxytocin has attracted great attention of the general public, basic neuroscience researchers, psychologists, and psychiatrists due to its profound pro-social, anxiolytic, and "anti-stress" behavioral and physiological effects, and its potential application for treatment of mental diseases associated with altered socio-emotional competence. During the last decade, substantial progress has been achieved in understanding the complex neurobiology of the oxytocin system, including oxytocinergic pathways, local release patterns, and oxytocin receptor distribution in the brain, as well as intraneuronal oxytocin receptor signaling. However, the picture of oxytocin actions remains far from being complete, and the central question remains: "How does a single neuropeptide exert such pleotropic actions?" Although this phenomenon, typical for many of about 100 identified neuropeptides, may emerge from the anatomical divergence of oxytocin neurons, their multiple central projections, distinct oxytocin-sensitive cell types in different brain regions, and multiple intraneuronal signaling pathways determining the specific cellular response, further basic studies are required. In conjunction, numerous reports on positive effects of intranasal application of oxytocin on human brain networks controlling socio-emotional behavior in health and disease require harmonic tandems of basic researchers and clinicians. During the COVID-19 crisis in 2020, oxytocin research seems central as question of social isolation-induced inactivation of the oxytocin system, and buffering effects of either activation of the endogenous system or intranasal application of synthetic oxytocin need to be thoroughly investigated.
Collapse
|
14
|
Affiliation(s)
- Yuri Bozzi
- Center for Mind/Brain Sciences (CIMeC), University of Trento, Italy; CNR Neuroscience Institute, Pisa, Italy
| | - Michela Fagiolini
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
15
|
Lefter R, Ciobica A, Antioch I, Ababei DC, Hritcu L, Luca AC. Oxytocin Differentiated Effects According to the Administration Route in a Prenatal Valproic Acid-Induced Rat Model of Autism. ACTA ACUST UNITED AC 2020; 56:medicina56060267. [PMID: 32485966 PMCID: PMC7353871 DOI: 10.3390/medicina56060267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/14/2020] [Accepted: 05/25/2020] [Indexed: 12/31/2022]
Abstract
Background and objectives: The hormone oxytocin (OXT) has already been reported in both human and animal studies for its promising therapeutic potential in autism spectrum disorder (ASD), but the comparative effectiveness of various administration routes, whether central or peripheral has been insufficiently studied. In the present study, we examined the effects of intranasal (IN) vs. intraperitoneal (IP) oxytocin in a valproic-acid (VPA) autistic rat model, focusing on cognitive and mood behavioral disturbances, gastrointestinal transit and central oxidative stress status. Materials and Methods: VPA prenatally-exposed rats (500 mg/kg; age 90 days) in small groups of 5 (n = 20 total) were given OXT by IP injection (10 mg/kg) for 8 days consecutively or by an adapted IN pipetting protocol (12 IU/kg, 20 μL/day) for 4 consecutive days. Behavioral tests were performed during the last three days of OXT treatment, and OXT was administrated 20 minutes before each behavioral testing for each rat. Biochemical determination of oxidative stress markers in the temporal area included superoxide dismutase (SOD), glutathione peroxidase (GPx) and malondialdehyde (MDA). A brief quantitative assessment of fecal discharge over a period of 24 hours was performed at the end of the OXT treatment to determine differences in intestinal transit. Results: OXT improved behavioral and oxidative stress status in both routes of administration, but IN treatment had significantly better outcome in improving short-term memory, alleviating depressive manifestations and mitigating lipid peroxidation in the temporal lobes. Significant correlations were also found between behavioral parameters and oxidative stress status in rats after OXT administration. The quantitative evaluation of the gastrointestinal (GI) transit indicated lower fecal pellet counts in the VPA group and homogenous average values for the control and both OXT treated groups. Conclusions: The data from the present study suggest OXT IN administration to be more efficient than IP injections in alleviating autistic cognitive and mood dysfunctions in a VPA-induced rat model. OXT effects on the cognitive and mood behavior of autistic rats may be associated with its effects on oxidative stress. Additionally, present results provide preliminary evidence that OXT may have a balancing effect on gastrointestinal motility.
Collapse
Affiliation(s)
- Radu Lefter
- Center of Biomedical Research, Romanian Academy, B dul Carol I, No 8, 700505 Iasi, Romania;
| | - Alin Ciobica
- Department of Research, Faculty of Biology, Alexandru Ioan Cuza University, B dul Carol I, No 11, 700506 Iasi, Romania;
- Correspondence: (A.C.); (L.H.)
| | - Iulia Antioch
- Department of Research, Faculty of Biology, Alexandru Ioan Cuza University, B dul Carol I, No 11, 700506 Iasi, Romania;
| | - Daniela Carmen Ababei
- “Grigore T.Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (D.C.A.); (A.-C.L.)
| | - Luminita Hritcu
- Faculty of Veterinary Medicine, University of Agricultural Sciencies and Veterinary Medicine “Ion Ionescu de la Brad” of Iasi, 3rd Mihail Sadoveanu Alley, 700490 Iasi, Romania
- Correspondence: (A.C.); (L.H.)
| | - Alina-Costina Luca
- “Grigore T.Popa” University of Medicine and Pharmacy, 16, Universitatii Street, 700115 Iasi, Romania; (D.C.A.); (A.-C.L.)
| |
Collapse
|