1
|
Wakuda T, Benner S, Uemura Y, Nishimura T, Kojima M, Kuroda M, Matsumoto K, Kanai C, Inada N, Harada T, Kameno Y, Munesue T, Inoue J, Umemura K, Yamauchi A, Ogawa N, Kushima I, Suyama S, Saito T, 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, Ozaki N, Kosaka H, Okada T, Kuwabara H, Yamasue H. Oxytocin-induced increases in cytokines and clinical effect on the core social features of autism: Analyses of RCT datasets. Brain Behav Immun 2024; 118:398-407. [PMID: 38461957 DOI: 10.1016/j.bbi.2024.03.013] [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: 09/18/2023] [Revised: 02/08/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024] Open
Abstract
Although oxytocin may provide a novel therapeutics for the core features of autism spectrum disorder (ASD), previous results regarding the efficacy of repeated or higher dose oxytocin are controversial, and the underlying mechanisms remain unclear. The current study is aimed to clarify whether repeated oxytocin alter plasma cytokine levels in relation to clinical changes of autism social core feature. Here we analyzed cytokine concentrations using comprehensive proteomics of plasmas of 207 adult males with high-functioning ASD collected from two independent multi-center large-scale randomized controlled trials (RCTs): Testing effects of 4-week intranasal administrations of TTA-121 (A novel oxytocin spray with enhanced bioavailability: 3U, 6U, 10U, or 20U/day) and placebo in the crossover discovery RCT; 48U/day Syntocinon or placebo in the parallel-group verification RCT. Among the successfully quantified 17 cytokines, 4 weeks TTA-121 6U (the peak dose for clinical effects) significantly elevated IL-7 (9.74, 95 % confidence interval [CI] 3.59 to 15.90, False discovery rate corrected P (PFDR) < 0.001), IL-9 (56.64, 20.46 to 92.82, PFDR < 0.001) and MIP-1b (18.27, 4.96 to 31.57, PFDR < 0.001) compared with placebo. Inverted U-shape dose-response relationships peaking at TTA-121 6U were consistently observed for all these cytokines (IL-7: P < 0.001; IL-9: P < 0.001; MIP-1b: P = 0.002). Increased IL-7 and IL-9 in participants with ASD after 4 weeks TTA-121 6U administration compared with placebo was verified in the confirmatory analyses in the dataset before crossover (PFDR < 0.001). Furthermore, the changes in all these cytokines during 4 weeks of TTA-121 10U administration revealed associations with changes in reciprocity score, the original primary outcome, observed during the same period (IL-7: Coefficient = -0.05, -0.10 to 0.003, P = 0.067; IL-9: -0.01, -0.02 to -0.003, P = 0.005; MIP-1b: -0.02, -0.04 to -0.007, P = 0.005). These findings provide the first evidence for a role of interaction between oxytocin and neuroinflammation in the change of ASD core social features, and support the potential role of this interaction as a novel therapeutic seed. Trial registration: UMIN000015264, NCT03466671/UMIN000031412.
Collapse
Affiliation(s)
- Tomoyasu Wakuda
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Seico Benner
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan; Center for Health and Environmental Risk Research, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, Ibaraki 305-8506, Japan
| | - Yukari Uemura
- Biostatistics Section, Department of Data Science, Center for Clinical Science, National Center for Global Health and Medicine, 1-21-1 Toyama, Shinjuku-ku, Tokyo 162-8655, Japan
| | - Tomoko Nishimura
- Department of Child Development, United Graduate School of Child Development at Hamamatsu, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Masaki Kojima
- Department of Child Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Miho Kuroda
- Department of Child Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kaori Matsumoto
- Graduate School of Psychology, Kanazawa Institute of Technology, 7-1 Ohgigaoka, Nonoichi, Ishikawa 921-8501, Japan
| | - Chieko Kanai
- Child Development and Education, Faculty of Humanities, Wayo Women's University, 2-3-1 Konodai, Ichikawa, Chiba 272-8533, Japan
| | - Naoko Inada
- Department of Psychology, Faculty of Liberal Arts, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
| | - Taeko Harada
- Department of Child Development, United Graduate School of Child Development at Hamamatsu, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Yosuke Kameno
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan; Department of Child Development, United Graduate School of Child Development at Hamamatsu, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Toshio Munesue
- Research Center for Child Mental Development, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Jun Inoue
- Department of Child and Adolescent Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Kazuo Umemura
- Department of Pharmacology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 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
| | - Satoshi Suyama
- Department of Child and Adolescent Psychiatry, Hokkaido University Hospital, Kita 14, Nishi 5, Kita-ku, Sapporo, Hokkaido 060-8648, Japan
| | - Takuya Saito
- Department of Child and Adolescent Psychiatry, Hokkaido University Hospital, Kita 14, Nishi 5, Kita-ku, Sapporo, Hokkaido 060-8648, Japan
| | - Junko Hamada
- Department of Child Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yukiko Kano
- Department of Child Neuropsychiatry, Graduate 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, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Saya Kikuchi
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Moe Seto
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Hiroaki Tomita
- Department of Psychiatry, Graduate School of Medicine, Tohoku University, 1-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8574, Japan
| | - Noriko Miyoshi
- Department of Psychiatry, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan; United Graduate School of Child Development, Osaka University, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Megumi Matsumoto
- Department of Psychiatry, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yuko Kawaguchi
- Department of Psychiatry, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Koji Kanai
- Department of Psychiatry, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Manabu Ikeda
- Department of Psychiatry, Osaka University Graduate School of Medicine, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan; United Graduate School of Child Development, Osaka University, 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Itta Nakamura
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Shuichi Isomura
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yoji Hirano
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; Department of Psychiatry, Division of Clinical Neuroscience, Faculty of Medicine, University of Miyazaki, 5200 Kiyotake-cho, Kihara, Miyazaki, Miyazaki 889-1692, Japan
| | - Toshiaki Onitsuka
- National Hospital Organization Sakakibara Hospital, 777 Sakakibara-cho, Tsu, Mie 514-1292, Japan
| | - Norio Ozaki
- Pathophysiology of Mental Disorders, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi 466-8550, Japan
| | - Hirotaka Kosaka
- Department of Neuropsychiatry, Faculty of Medical Sciences, University of Fukui, 23-3 Matsuoka, Shimoaizuki, Eiheiji-cho, Yoshida-gun, 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
| | - Hitoshi Kuwabara
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan; Department of Child Development, United Graduate School of Child Development at Hamamatsu, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Hidenori Yamasue
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan; Department of Child Development, United Graduate School of Child Development at Hamamatsu, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan.
| |
Collapse
|
2
|
Rahmatinia M, Mohseni-Bandpei A, Khodagholi F, Abdollahifar MA, Amouei Torkmahalleh M, Hassani Moghaddam M, Hopke PK, Ghavimehr E, Bazzazpour S, Shahsavani A. Exposure to different PM 2.5 extracts induces gliosis and changes behavior in male rats similar to autism spectrum disorders features. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 340:122804. [PMID: 37907193 DOI: 10.1016/j.envpol.2023.122804] [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: 07/31/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/02/2023]
Abstract
Epidemiological studies have documented that exposure to fine particulate matter (PM2.5) could affect neurodevelopment, thereby leading to autism spectrum disorders (ASD). Nevertheless, there is little laboratory data to support this epidemiological evidence. In the current study, we carried out a series of experiments to assess whether developmental exposures to different extracts of PM2.5 can result in ASD-like behavioral, biochemical, and immunohistochemical characteristics in male rat offspring. PM2.5 samples were collected daily for a year, and monthly composites were extracted with an acetone-hexane mixture. The extracts were analyzed for their chemical constituents. Three groups of rats were exposed to the different PM2.5 extracts during pre- and postnatal periods. All exposed groups of rats exhibited typical behavioral features of ASD, including increased repetitive and depression-related behaviors. We also found microglia and astrocytes activation and decreased concentrations of oxytocin (OXT) in the brain regions of exposed rats compared with control rats. Comparing the current results with a prior study, the induced biological effects followed a sequence of whole particles of PM2.5 > organic extract > inorganic extract. These findings indicated that exposure to PM2.5 can elicit ASD-like features in rats and raise concerns about particulate matter as a possible trigger for the induction of ASD in humans; therefore, mitigating the contents of the PAHs and metals could reduce the PM2.5 neurotoxicity.
Collapse
Affiliation(s)
- Masoumeh Rahmatinia
- Department of Environmental Health Engineering, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Air Quality and Climate Change Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Anoushiravan Mohseni-Bandpei
- Department of Environmental Health Engineering, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Air Quality and Climate Change Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Amouei Torkmahalleh
- Division of Environmental and Occupational Health Sciences, School of Public Health, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Philip K Hopke
- Department of Public Health Sciences, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Ehsan Ghavimehr
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Shahriyar Bazzazpour
- Department of Environmental Health Engineering, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Shahsavani
- Department of Environmental Health Engineering, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Air Quality and Climate Change Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Yamasue H. Is the efficacy of oxytocin for autism diminished at higher dosages or repeated doses?: Potential mechanisms and candidate solutions. Peptides 2024; 171:171133. [PMID: 38072084 DOI: 10.1016/j.peptides.2023.171133] [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: 11/03/2023] [Revised: 12/05/2023] [Accepted: 12/05/2023] [Indexed: 12/17/2023]
Abstract
No approved pharmacological intervention currently exists to address the core symptoms of autism spectrum disorder, a prevalent neurodevelopmental condition. However, there is a growing body of empirical evidence highlighting oxytocin's modulatory effects on social and communicative behaviors. Numerous single-dose trials have consistently demonstrated the efficacy of oxytocin in ameliorating behavioral and neural measurements associated with the core symptoms of autism spectrum disorder. Nevertheless, prior investigations involving the repeated administration of oxytocin have yielded disparate findings concerning its effectiveness, particularly in relation to clinical measures of the core symptoms of autism spectrum disorder. Recent studies have also raised the possibility of diminishing efficacy of oxytocin over time, particularly when higher or recurrent dosages of oxytocin are administered. This review article aims to provide an overview of previous studies examining this issue. Furthermore, it aims to discuss the potential mechanisms underlying these effects, including the interaction between oxytocin and vasopressin, as well as potential strategies for addressing the challenges mentioned. This review's overall objective is to provide insights into the potential development of innovative therapeutics to mitigate the core symptoms of autism spectrum disorder, representing potential breakthroughs in the treatment of this complex neurodevelopmental condition.
Collapse
Affiliation(s)
- Hidenori Yamasue
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka 431-3192, Japan.
| |
Collapse
|
4
|
Rahmati-Holasoo H, Salek Maghsoudi A, Akbarzade M, Gholami M, Shadboorestan A, Vakhshiteh F, Armandeh M, Hassani S. Oxytocin protective effects on zebrafish larvae models of autism-like spectrum disorder. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:316-325. [PMID: 36865037 PMCID: PMC9922369 DOI: 10.22038/ijbms.2023.68165.14889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/02/2023] [Indexed: 03/04/2023]
Abstract
Objectives Autism is a complicated neurodevelopmental disorder characterized by social interaction deficiencies, hyperactivity, anxiety, communication disorders, and a limited range of interests. The zebrafish (Danio rerio) is a social vertebrate used as a biomedical research model to understand social behavior mechanisms. Materials and Methods After spawning, the eggs were exposed to sodium valproate for 48 hr, after which the eggs were divided into eight groups. Except for the positive and control groups, there were six treatment groups based on oxytocin concentration (25, 50, and 100 μM) and time point (24 and 48 hr). Treatment was performed on days 6 and 7, examined by labeling oxytocin with fluorescein-5-isothiocyanate (FITC) and imaging with confocal microscopy and the expression levels of potential genes associated with the qPCR technique. Behavioral studies, including light-dark background preference test, shoaling behavior, mirror test, and social preference, were performed on 10, 11, 12, and 13 days post fertilization (dpf), respectively. Results The results showed that the most significant effect of oxytocin was at the concentration of 50 μM and the time point of 48 hr. Increased expression of shank3a, shank3b, and oxytocin receptor genes was also significant at this oxytocin concentration. Light-dark background preference results showed that oxytocin in the concentration of 50 µM significantly increased the number of crosses between dark and light areas compared with valproic acid (positive group). Also, oxytocin showed an increase in the frequency and time of contact between the two larvae. We showed a decrease in the distance in the larval group and an increase in time spent at a distance of one centimeter from the mirror. Conclusion Our findings showed that the increased gene expression of shank3a, shank3b, and oxytocin receptors improved autistic behavior. Based on this study some indications showed that oxytocin administration in the larval stage could significantly improve the autism-like spectrum.
Collapse
Affiliation(s)
- Hooman Rahmati-Holasoo
- Department of Aquatic Animal Health, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran, Center of Excellence for Warm Water Fish Health and Disease, Shahid Chamran University of Ahvaz, Ahvaz, Iran,These authors contributed eqully to this work
| | - Armin Salek Maghsoudi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran,These authors contributed eqully to this work
| | - Milad Akbarzade
- Department of Aquatic Animal Health, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mahdi Gholami
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Amir Shadboorestan
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Faezeh Vakhshiteh
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Maryam Armandeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Shokoufeh Hassani
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran,Corresponding author: Shokoufeh Hassani, Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| |
Collapse
|
5
|
Olszewski PK, Noble EE, Paiva L, Ueta Y, Blevins JE. Oxytocin as a potential pharmacological tool to combat obesity. J Neuroendocrinol 2022; 34:e13106. [PMID: 35192207 PMCID: PMC9372234 DOI: 10.1111/jne.13106] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/06/2022] [Accepted: 02/08/2022] [Indexed: 11/30/2022]
Abstract
The neuropeptide oxytocin (OT) has emerged as an important anorexigen in the regulation of food intake and energy balance. It has been shown that the release of OT and activation of hypothalamic OT neurons coincide with food ingestion. Its effects on feeding have largely been attributed to limiting meal size through interactions in key regulatory brain regions governing the homeostatic control of food intake such as the hypothalamus and hindbrain in addition to key feeding reward areas such as the nucleus accumbens and ventral tegmental area. Furthermore, the magnitude of an anorexigenic response to OT and feeding-related activation of the brain OT circuit are modified by the composition and flavor of a diet, as well as by a social context in which a meal is consumed. OT is particularly effective in reducing consumption of carbohydrates and sweet tastants. Pharmacologic, genetic, and pair-feeding studies indicate that OT-elicited weight loss cannot be fully explained by reductions of food intake and that the overall impact of OT on energy balance is also partly a result of OT-elicited changes in lipolysis, energy expenditure, and glucose regulation. Peripheral administration of OT mimics many of its effects when it is given into the central nervous system, raising the questions of whether and to what extent circulating OT acts through peripheral OT receptors to regulate energy balance. Although OT has been found to elicit weight loss in female mice, recent studies have indicated that sex and estrous cycle may impact oxytocinergic modulation of food intake. Despite the overall promising basic research data, attempts to use OT in the clinical setting to combat obesity and overeating have generated somewhat mixed results. The focus of this mini-review is to briefly summarize the role of OT in feeding and metabolism, address gaps and inconsistencies in our knowledge, and discuss some of the limitations to the potential use of chronic OT that should help guide future research on OT as a tailor-made anti-obesity therapeutic.
Collapse
Affiliation(s)
- Pawel K Olszewski
- Faculty of Science and Engineering, University of Waikato, Waikato, New Zealand
- Department of Food Science and Nutrition, College of Food, Agricultural and Natural Resource Sciences, University of Minnesota, St Paul, Minnesota, USA
- Department of Integrative Biology and Physiology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Emily E Noble
- Department of Nutritional Sciences, University of Georgia, Athens, Georgia, USA
| | - Luis Paiva
- Instituto de Ciencia Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Yoichi Ueta
- Department of Physiology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Fukuoka, Japan
| | - James E Blevins
- Department of Veterans Affairs Medical Center, VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Seattle, Washington, USA
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
6
|
Wei D, Tsheringla S, McPartland JC, Allsop AZASA. Combinatorial approaches for treating neuropsychiatric social impairment. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210051. [PMID: 35858103 PMCID: PMC9274330 DOI: 10.1098/rstb.2021.0051] [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: 10/01/2021] [Accepted: 05/13/2022] [Indexed: 01/30/2023] Open
Abstract
Social behaviour is an essential component of human life and deficits in social function are seen across multiple psychiatric conditions with high morbidity. However, there are currently no FDA-approved treatments for social dysfunction. Since social cognition and behaviour rely on multiple signalling processes acting in concert across various neural networks, treatments aimed at social function may inherently require a combinatorial approach. Here, we describe the social neurobiology of the oxytocin and endocannabinoid signalling systems as well as translational evidence for their use in treating symptoms in the social domain. We leverage this systems neurobiology to propose a network-based framework that involves pharmacology, psychotherapy, non-invasive brain stimulation and social skills training to combinatorially target trans-diagnostic social impairment. Lastly, we discuss the combined use of oxytocin and endocannabinoids within our proposed framework as an illustrative strategy to treat specific aspects of social function. Using this framework provides a roadmap for actionable treatment strategies for neuropsychiatric social impairment. This article is part of the theme issue 'Interplays between oxytocin and other neuromodulators in shaping complex social behaviours'.
Collapse
Affiliation(s)
- Don Wei
- Department of Psychiatry, UCLA, Los Angeles, CA, USA
| | | | | | | |
Collapse
|
7
|
McParlin Z, Cerritelli F, Rossettini G, Friston KJ, Esteves JE. Therapeutic Alliance as Active Inference: The Role of Therapeutic Touch and Biobehavioural Synchrony in Musculoskeletal Care. Front Behav Neurosci 2022; 16:897247. [PMID: 35846789 PMCID: PMC9280207 DOI: 10.3389/fnbeh.2022.897247] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/24/2022] [Indexed: 12/05/2022] Open
Abstract
Touch is recognised as crucial for survival, fostering cooperative communication, accelerating recovery, reducing hospital stays, and promoting overall wellness and the therapeutic alliance. In this hypothesis and theory paper, we present an entwined model that combines touch for alignment and active inference to explain how the brain develops "priors" necessary for the health care provider to engage with the patient effectively. We appeal to active inference to explain the empirically integrative neurophysiological and behavioural mechanisms that underwrite synchronous relationships through touch. Specifically, we offer a formal framework for understanding - and explaining - the role of therapeutic touch and hands-on care in developing a therapeutic alliance and synchrony between health care providers and their patients in musculoskeletal care. We first review the crucial importance of therapeutic touch and its clinical role in facilitating the formation of a solid therapeutic alliance and in regulating allostasis. We then consider how touch is used clinically - to promote cooperative communication, demonstrate empathy, overcome uncertainty, and infer the mental states of others - through the lens of active inference. We conclude that touch plays a crucial role in achieving successful clinical outcomes and adapting previous priors to create intertwined beliefs. The ensuing framework may help healthcare providers in the field of musculoskeletal care to use hands-on care to strengthen the therapeutic alliance, minimise prediction errors (a.k.a., free energy), and thereby promote recovery from physical and psychological impairments.
Collapse
Affiliation(s)
- Zoe McParlin
- Clinical-Based Human Research Department, Foundation COME Collaboration, Pescara, Italy
| | - Francesco Cerritelli
- Clinical-Based Human Research Department, Foundation COME Collaboration, Pescara, Italy
| | | | - Karl J. Friston
- Institute of Neurology, Wellcome Centre for Human Neuroimaging, London, United Kingdom
| | - Jorge E. Esteves
- Clinical-Based Human Research Department, Foundation COME Collaboration, Pescara, Italy
- Malta ICOM Educational, Gzira, Malta
- University College of Osteopathy, London, United Kingdom
| |
Collapse
|
8
|
McParlin Z, Cerritelli F, Friston KJ, Esteves JE. Therapeutic Alliance as Active Inference: The Role of Therapeutic Touch and Synchrony. Front Psychol 2022; 13:783694. [PMID: 35250723 PMCID: PMC8892201 DOI: 10.3389/fpsyg.2022.783694] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 01/19/2022] [Indexed: 12/15/2022] Open
Abstract
Recognizing and aligning individuals' unique adaptive beliefs or "priors" through cooperative communication is critical to establishing a therapeutic relationship and alliance. Using active inference, we present an empirical integrative account of the biobehavioral mechanisms that underwrite therapeutic relationships. A significant mode of establishing cooperative alliances-and potential synchrony relationships-is through ostensive cues generated by repetitive coupling during dynamic touch. Established models speak to the unique role of affectionate touch in developing communication, interpersonal interactions, and a wide variety of therapeutic benefits for patients of all ages; both neurophysiologically and behaviorally. The purpose of this article is to argue for the importance of therapeutic touch in establishing a therapeutic alliance and, ultimately, synchrony between practitioner and patient. We briefly overview the importance and role of therapeutic alliance in prosocial and clinical interactions. We then discuss how cooperative communication and mental state alignment-in intentional communication-are accomplished using active inference. We argue that alignment through active inference facilitates synchrony and communication. The ensuing account is extended to include the role of (C-) tactile afferents in realizing the beneficial effect of therapeutic synchrony. We conclude by proposing a method for synchronizing the effects of touch using the concept of active inference.
Collapse
Affiliation(s)
- Zoe McParlin
- Foundation COME Collaboration, Clinical-Based Human Research Department, Pescara, Italy
| | - Francesco Cerritelli
- Foundation COME Collaboration, Clinical-Based Human Research Department, Pescara, Italy
| | - Karl J. Friston
- Wellcome Centre for Human Neuroimaging, Institute of Neurology, London, United Kingdom
| | - Jorge E. Esteves
- Foundation COME Collaboration, Clinical-Based Human Research Department, Pescara, Italy
- Malta ICOM Educational Ltd., Gzira, Malta
- Research Department, University College of Osteopathy, London, United Kingdom
| |
Collapse
|
9
|
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: 7.0] [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
|
10
|
Yu H, Miao W, Ji E, Huang S, Jin S, Zhu X, Liu MZ, Sun YG, Xu F, Yu X. Social touch-like tactile stimulation activates a tachykinin 1-oxytocin pathway to promote social interactions. Neuron 2022; 110:1051-1067.e7. [PMID: 35045339 DOI: 10.1016/j.neuron.2021.12.022] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 08/29/2021] [Accepted: 12/15/2021] [Indexed: 12/21/2022]
Abstract
It is well known that affective and pleasant touch promotes individual well-being and facilitates affiliative social communication, although the neural circuit that mediates this process is largely unknown. Here, we show that social-touch-like tactile stimulation (ST) enhances firing of oxytocin neurons in the mouse paraventricular hypothalamus (PVH) and promotes social interactions and positively reinforcing place preference. These results link pleasant somatosensory stimulation to increased social interactions and positive affective valence. We further show that tachykinin 1 (Tac1+) neurons in the lateral and ventrolateral periaqueductal gray (l/vlPAG) send monosynaptic excitatory projections to PVH oxytocin neurons. Functionally, activation of PVH-projecting Tac1+ neurons increases firing of oxytocin neurons, promotes social interactions, and increases preference for the social touch context, whereas reducing activity of Tac1+ neurons abolishes ST-induced oxytocin neuronal firing. Together, these results identify a dipeptidergic pathway from l/vlPAG Tac1+ neurons to PVH oxytocin neurons, through which pleasant sensory experience promotes social behavior.
Collapse
Affiliation(s)
- Hang Yu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wanying Miao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - En Ji
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shajin Huang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Sen Jin
- Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Xutao Zhu
- Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Ming-Zhe Liu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan-Gang Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fuqiang Xu
- University of Chinese Academy of Sciences, Beijing 100049, China; Brain Cognition and Brain Disease Institute, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Xiang Yu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and Peking University McGovern Institute, Peking University, Beijing 100871, China; Autism Research Center of Peking University Health Science Center, Beijing 100191, China; Chinese Institute for Brain Research, Beijing 102206, China.
| |
Collapse
|
11
|
Leptin enhances social motivation and reverses chronic unpredictable stress-induced social anhedonia during adolescence. Mol Psychiatry 2022; 27:4948-4958. [PMID: 36138127 PMCID: PMC9763124 DOI: 10.1038/s41380-022-01778-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/12/2022] [Accepted: 09/02/2022] [Indexed: 01/19/2023]
Abstract
Social anhedonia, a loss of interest and pleasure in social interactions, is a common symptom of major depression as well as other psychiatric disorders. Depression can occur at any age, but typically emerges in adolescence or early adulthood, which represents a sensitive period for social interaction that is vulnerable to stress. In this study, we evaluated social interaction reward using a conditioned place preference (CPP) paradigm in adolescent male and female mice. Adolescent mice of both sexes exhibited a preference for the social interaction-associated context. Chronic unpredictable stress (CUS) impaired the development of CPP for social interaction, mimicking social anhedonia in depressed adolescents. Conversely, administration of leptin, an adipocyte-derived hormone, enhanced social interaction-induced CPP in non-stressed control mice and reversed social anhedonia in CUS mice. By dissecting the motivational processes of social CPP into social approach and isolation avoidance components, we demonstrated that leptin treatment increased isolation aversion without overt social reward effect. Further mechanistic exploration revealed that leptin stimulated oxytocin gene transcription in the paraventricular nucleus of the hypothalamus, while oxytocin receptor blockade abolished the leptin-induced enhancement of socially-induced CPP. These results establish that chronic unpredictable stress can be used to study social anhedonia in adolescent mice and provide evidence that leptin modulates social motivation possibly via increasing oxytocin synthesis and oxytocin receptor activation.
Collapse
|
12
|
Niu J, Tong J, Blevins JE. Oxytocin as an Anti-obesity Treatment. Front Neurosci 2021; 15:743546. [PMID: 34720864 PMCID: PMC8549820 DOI: 10.3389/fnins.2021.743546] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 09/16/2021] [Indexed: 12/19/2022] Open
Abstract
Obesity is a growing health concern, as it increases risk for heart disease, hypertension, type 2 diabetes, cancer, COVID-19 related hospitalizations and mortality. However, current weight loss therapies are often associated with psychiatric or cardiovascular side effects or poor tolerability that limit their long-term use. The hypothalamic neuropeptide, oxytocin (OT), mediates a wide range of physiologic actions, which include reproductive behavior, formation of prosocial behaviors and control of body weight. We and others have shown that OT circumvents leptin resistance and elicits weight loss in diet-induced obese rodents and non-human primates by reducing both food intake and increasing energy expenditure (EE). Chronic intranasal OT also elicits promising effects on weight loss in obese humans. This review evaluates the potential use of OT as a therapeutic strategy to treat obesity in rodents, non-human primates, and humans, and identifies potential mechanisms that mediate this effect.
Collapse
Affiliation(s)
- JingJing Niu
- VA Puget Sound Health Care System, Office of Research and Development, Medical Research Service, Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| | - Jenny Tong
- VA Puget Sound Health Care System, Office of Research and Development, Medical Research Service, Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| | - James E. Blevins
- VA Puget Sound Health Care System, Office of Research and Development, Medical Research Service, Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
13
|
Oxytocin Is a Positive Allosteric Modulator of κ-Opioid Receptors but Not δ-Opioid Receptors in the G Protein Signaling Pathway. Cells 2021; 10:cells10102651. [PMID: 34685631 PMCID: PMC8534029 DOI: 10.3390/cells10102651] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 09/29/2021] [Accepted: 09/29/2021] [Indexed: 01/22/2023] Open
Abstract
Oxytocin (OT) influences various physiological functions such as uterine contractions, maternal/social behavior, and analgesia. Opioid signaling pathways are involved in one of the analgesic mechanisms of OT. We previously showed that OT acts as a positive allosteric modulator (PAM) and enhances μ-opioid receptor (MOR) activity. In this study, which focused on other opioid receptor (OR) subtypes, we investigated whether OT influences opioid signaling pathways as a PAM for δ-OR (DOR) or κ-OR (KOR) using human embryonic kidney-293 cells expressing human DOR or KOR, respectively. The CellKeyTM results showed that OT enhanced impedance induced by endogenous/exogenous KOR agonists on KOR-expressing cells. OT did not affect DOR activity induced by endogenous/exogenous DOR agonists. OT potentiated the KOR agonist-induced Gi/o protein-mediated decrease in intracellular cAMP, but did not affect the increase in KOR internalization caused by the KOR agonists dynorphin A and (-)-U-50488 hydrochloride (U50488). OT did not bind to KOR orthosteric binding sites and did not affect the binding affinities of dynorphin A and U50488 for KOR. These results suggest that OT is a PAM of KOR and MOR and enhances G protein signaling without affecting β-arrestin signaling. Thus, OT has potential as a specific signaling-biased PAM of KOR.
Collapse
|
14
|
Oxytocin, Erectile Function and Sexual Behavior: Last Discoveries and Possible Advances. Int J Mol Sci 2021; 22:ijms221910376. [PMID: 34638719 PMCID: PMC8509000 DOI: 10.3390/ijms221910376] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 12/30/2022] Open
Abstract
A continuously increasing amount of research shows that oxytocin is involved in numerous central functions. Among the functions in which oxytocin is thought to be involved are those that play a role in social and sexual behaviors, and the involvement of central oxytocin in erectile function and sexual behavior was indeed one of the first to be discovered in laboratory animals in the 1980s. The first part of this review summarizes the results of studies done in laboratory animals that support a facilitatory role of oxytocin in male and female sexual behavior and reveal mechanisms through which this ancient neuropeptide participates in concert with other neurotransmitters and neuropeptides in this complex function, which is fundamental for the species reproduction. The second part summarizes the results of studies done mainly with intranasal oxytocin in men and women with the aim to translate the results found in laboratory animals to humans. Unexpectedly, the results of these studies do not appear to confirm the facilitatory role of oxytocin found in male and female sexual behavior in animals, both in men and women. Possible explanations for the failure of oxytocin to improve sexual behavior in men and women and strategies to attempt to overcome this impasse are considered.
Collapse
|
15
|
Zhang J, Li SJ, Miao W, Zhang X, Zheng JJ, Wang C, Yu X. Oxytocin Regulates Synaptic Transmission in the Sensory Cortices in a Developmentally Dynamic Manner. Front Cell Neurosci 2021; 15:673439. [PMID: 34177467 PMCID: PMC8221398 DOI: 10.3389/fncel.2021.673439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 05/10/2021] [Indexed: 01/08/2023] Open
Abstract
The development and stabilization of neuronal circuits are critical to proper brain function. Synapses are the building blocks of neural circuits. Here we examine the effects of the neuropeptide oxytocin on synaptic transmission in L2/3 pyramidal neurons of the barrel field of the primary somatosensory cortex (S1BF). We find that perfusion of oxytocin onto acute brain slices significantly increases the frequency of miniature excitatory postsynaptic currents (mEPSC) of S1BF L2/3 pyramidal neurons at P10 and P14, but reduces it at the later ages of P22 and P28; the transition occurs at around P18. Since oxytocin expression is itself regulated by sensory experience, we also examine whether the effects of oxytocin on excitatory synaptic transmission correlate with that of sensory experience. We find that, indeed, the effects of sensory experience and oxytocin on excitatory synaptic transmission of L2/3 pyramidal neurons both peak at around P14 and plateau around P18, suggesting that they regulate a specific form of synaptic plasticity in L2/3 pyramidal neurons, with a sensitive/critical period ending around P18. Consistently, oxytocin receptor (Oxtr) expression in glutamatergic neurons of the upper layers of the cerebral cortex peaks around P14. By P28, however, Oxtr expression becomes more prominent in GABAergic neurons, especially somatostatin (SST) neurons. At P28, oxytocin perfusion increases inhibitory synaptic transmission and reduces excitatory synaptic transmission, effects that result in a net reduction of neuronal excitation, in contrast to increased excitation at P14. Using oxytocin knockout mice and Oxtr conditional knockout mice, we show that loss-of-function of oxytocin affects baseline excitatory synaptic transmission, while Oxtr is required for oxytocin-induced changes in excitatory synaptic transmission, at both P14 and P28. Together, these results demonstrate that oxytocin has complex and dynamic functions in regulating synaptic transmission in cortical L2/3 pyramidal neurons. These findings add to existing knowledge of the function of oxytocin in regulating neural circuit development and plasticity.
Collapse
Affiliation(s)
- Jing Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,Department of Clinical Pharmacology, Pharmacy College, Nanjing Medical University, Nanjing, China
| | - Shu-Jing Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Wanying Miao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xiaodi Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jing-Jing Zheng
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Chen Wang
- CAS Key Laboratory of Biological Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Xiang Yu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.,School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and Peking University McGovern Institute, Peking University, Beijing, China
| |
Collapse
|
16
|
Kato Y, Kuwabara H, Okada T, Munesue T, Benner S, Kuroda M, Kojima M, Yassin W, Eriguchi Y, Kameno Y, Murayama C, Nishimura T, Tsuchiya K, Kasai K, Ozaki N, Kosaka H, Yamasue H. Oxytocin-induced increase in N,N-dimethylglycine and time course of changes in oxytocin efficacy for autism social core symptoms. Mol Autism 2021; 12:15. [PMID: 33622389 PMCID: PMC7903697 DOI: 10.1186/s13229-021-00423-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 02/12/2021] [Indexed: 11/12/2022] Open
Abstract
Background Oxytocin is expected as a novel therapeutic agent for autism spectrum disorder (ASD) core symptoms. However, previous results on the efficacy of repeated administrations of oxytocin are controversial. Recently, we reported time-course changes in the efficacy of the neuropeptide underlying the controversial effects of repeated administration; however, the underlying mechanisms remained unknown. Methods The current study explored metabolites representing the molecular mechanisms of oxytocin’s efficacy using high-throughput metabolomics analysis on plasma collected before and after 6-week repeated intranasal administration of oxytocin (48 IU/day) or placebo in adult males with ASD (N = 106) who participated in a multi-center, parallel-group, double-blind, placebo-controlled, randomized controlled trial. Results Among the 35 metabolites measured, a significant increase in N,N-dimethylglycine was detected in the subjects administered oxytocin compared with those given placebo at a medium effect size (false discovery rate (FDR) corrected P = 0.043, d = 0.74, N = 83). Furthermore, subgroup analyses of the participants displaying a prominent time-course change in oxytocin efficacy revealed a significant effect of oxytocin on N,N-dimethylglycine levels with a large effect size (PFDR = 0.004, d = 1.13, N = 60). The increase in N,N-dimethylglycine was significantly correlated with oxytocin-induced clinical changes, assessed as changes in quantifiable characteristics of autistic facial expression, including both of improvements between baseline and 2 weeks (PFDR = 0.006, r = − 0.485, N = 43) and deteriorations between 2 and 4 weeks (PFDR = 0.032, r = 0.415, N = 37). Limitations The metabolites changes caused by oxytocin administration were quantified using peripheral blood and therefore may not directly reflect central nervous system changes. Conclusion Our findings demonstrate an association of N,N-dimethylglycine upregulation with the time-course change in the efficacy of oxytocin on autistic social deficits. Furthermore, the current findings support the involvement of the N-methyl-D-aspartate receptor and neural plasticity to the time-course change in oxytocin’s efficacy. Trial registration: A multi-center, parallel-group, placebo-controlled, double-blind, confirmatory trial of intranasal oxytocin in participants with autism spectrum disorders (the date registered: 30 October 2014; UMIN Clinical Trials Registry: https://upload.umin.ac.jp/cgi-open-bin/ctr_e/ctr_view.cgi?recptno=R000017703) (UMIN000015264).
Collapse
Affiliation(s)
- Yasuhiko Kato
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu City, 431-3192, Japan
| | - Hitoshi Kuwabara
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu City, 431-3192, Japan
| | - Takashi Okada
- Department of Psychiatry, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Toshio Munesue
- Research Center for Child Mental Development, Kanazawa University, 13-1 Takara-machi, Kanazawa, 920-8640, Japan
| | - Seico Benner
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu City, 431-3192, Japan
| | - Miho Kuroda
- Department of Child Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Masaki Kojima
- Department of Child Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Walid Yassin
- Department of Child Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yosuke Eriguchi
- Department of Child Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yosuke Kameno
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu City, 431-3192, Japan
| | - Chihiro Murayama
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu City, 431-3192, Japan
| | - Tomoko Nishimura
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Osaka/Kanazawa/Hamamatsu/Chiba/Fukui, Japan
| | - Kenji Tsuchiya
- United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Osaka/Kanazawa/Hamamatsu/Chiba/Fukui, Japan
| | - Kiyoto Kasai
- Department of Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Hirotaka Kosaka
- Department of Neuropsychiatry, Faculty of Medical Sciences, University of Fukui, Eiheiji, Fukui, 910-1193, Japan
| | - Hidenori Yamasue
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu City, 431-3192, Japan. .,United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Osaka/Kanazawa/Hamamatsu/Chiba/Fukui, Japan.
| |
Collapse
|
17
|
Benner S, Aoki Y, Watanabe T, Endo N, Abe O, Kuroda M, Kuwabara H, Kawakubo Y, Takao H, Kunimatsu A, Kasai K, Bito H, Kakeyama M, Yamasue H. Neurochemical evidence for differential effects of acute and repeated oxytocin administration. Mol Psychiatry 2021; 26:710-720. [PMID: 30262887 DOI: 10.1038/s41380-018-0249-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/08/2018] [Accepted: 08/08/2018] [Indexed: 12/18/2022]
Abstract
A discrepancy in oxytocin's behavioral effects between acute and repeated administrations indicates distinct underlying neurobiological mechanisms. The current study employed a combination of human clinical trial and animal study to compare neurochemical changes induced by acute and repeated oxytocin administrations. Human study analyzed medial prefrontal metabolite levels by using 1H-magnetic resonance spectroscopy, a secondary outcome in our randomized, double-blind, placebo-controlled crossover trial of 6 weeks intranasal administrations of oxytocin (48 IU/day) and placebo within-subject design in 17 psychotropic-free high-functioning men with autism spectrum disorder. Medial prefrontal transcript expression levels were analyzed in adult male C57BL/6J mice after intraperitoneal injection of oxytocin or saline either once (200 ng/100 μL/mouse, n = 12) or for 14 consecutive days (200 ng/100 μL/mouse/day, n = 16). As the results, repeated administration of oxytocin significantly decreased the medial prefrontal N-acetylaspartate (NAA; p = 0.043) and glutamate-glutamine levels (Glx; p = 0.001), unlike the acute oxytocin. The decreases were inversely and specifically associated (r = 0.680, p = 0.004 for NAA; r = 0.491, p = 0.053 for Glx) with oxytocin-induced improvements of medial prefrontal functional MRI activity during a social judgment task not with changes during placebo administrations. In wild-type mice, we found that repeated oxytocin administration reduced medial frontal transcript expression of N-methyl-D-aspartate receptor type 2B (p = 0.018), unlike the acute oxytocin, which instead changed the transcript expression associated with oxytocin (p = 0.0004) and neural activity (p = 0.0002). The present findings suggest that the unique sensitivity of the glutamatergic system to repeated oxytocin administration may explain the differential behavioral effects of oxytocin between acute and repeated administration.
Collapse
Affiliation(s)
- Seico Benner
- Department of Child Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yuta Aoki
- Department of Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takamitsu Watanabe
- Department of Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.,Institute of Cognitive Neuroscience, University College London, 17 Queen Square, London, WC1N 3AZ, United Kingdom
| | - Nozomi Endo
- Laboratory for Environmental Brain Science, Faculty of Human Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa, 359-1192, Japan.,Department of Anatomy and Cell Biology, Nara Medical University, Kashihara, Nara, 634-8521, Japan
| | - Osamu Abe
- Department of Radiology, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Miho Kuroda
- 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, Higashi-ku, Hamamatsu, 431-3192, Japan
| | - Yuki Kawakubo
- Department of Child Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hidemasa Takao
- Department of Radiology, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Akira Kunimatsu
- Department of Radiology, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kiyoto Kasai
- Department of Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Haruhiko Bito
- Department of Neurochemistry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Masaki Kakeyama
- Laboratory for Environmental Brain Science, Faculty of Human Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa, 359-1192, Japan
| | - Hidenori Yamasue
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, 431-3192, Japan.
| |
Collapse
|
18
|
Tanioka D, Chikahisa S, Shimizu N, Shiuchi T, Sakai N, Nishino S, Séi H. Intracranial mast cells contribute to the control of social behavior in male mice. Behav Brain Res 2021; 403:113143. [PMID: 33516739 DOI: 10.1016/j.bbr.2021.113143] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/13/2021] [Accepted: 01/22/2021] [Indexed: 12/16/2022]
Abstract
Mast cells (MCs) exist intracranially and have been reported to affect higher brain functions in rodents. However, the role of MCs in the regulation of emotionality and social behavior is unclear. In the present study, using male mice, we examined the relationship between MCs and social behavior and investigated the underlying mechanisms. Wild-type male mice intraventricularly injected with a degranulator of MCs exhibited a marked increase in a three-chamber sociability test. In addition, removal of MCs in Mast cell-specific Toxin Receptor-mediated Conditional cell Knock out (Mas-TRECK) male mice showed reduced social preference levels in a three-chamber sociability test without other behavioral changes, such as anxiety-like and depression-like behavior. Mas-TRECK male mice also had reduced serotonin content and serotonin receptor expression and increased oxytocin receptor expression in the brain. These results suggested that MCs may contribute to the regulation of social behavior in male mice. This effect may be partially mediated by serotonin derived from MCs in the brain.
Collapse
Affiliation(s)
- Daisuke Tanioka
- Department of Integrative Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Sachiko Chikahisa
- Department of Integrative Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Noriyuki Shimizu
- Department of Integrative Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Tetsuya Shiuchi
- Department of Integrative Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Noriaki Sakai
- Sleep & Circadian Neurobiology Laboratory, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Seiji Nishino
- Sleep & Circadian Neurobiology Laboratory, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Hiroyoshi Séi
- Department of Integrative Physiology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
19
|
Kumar R, Gulati M, Singh SK, Sharma D, Porwal O. Road From Nose to Brain for Treatment of Alzheimer: The Bumps and Humps. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 19:663-675. [PMID: 32640969 DOI: 10.2174/1871527319666200708124726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/15/2020] [Accepted: 04/27/2020] [Indexed: 11/22/2022]
Abstract
Vulnerability of the brain milieu to even the subtle changes in its normal physiology is guarded by a highly efficient blood brain barrier. A number of factors i.e. molecular weight of the drug, its route of administration, lipophilic character, etc. play a significant role in its sojourn through the Blood Brain Barrier (BBB) and limit the movement of drug into brain tissue through BBB. To overcome these problems, alternative routes of drug administration have been explored to target the drugs to brain tissue. Nasal route has been widely reported for the administration of drugs for treatment of Alzheimer. In this innovative approach, the challenge of BBB is bypassed. Through this route, both the larger as well as polar molecules can be made to reach the brain tissues. Generally, these systems are either pH dependent or temperature dependent. The present review highlights the anatomy of nose, mechanisms of drug delivery from nose to brain, critical factors in the formulation of nasal drug delivery system, nasal formulations of various drugs that have been tried for their nasal delivery for treatment of Alzheimer. It also dives deep to understand the factors that contribute to the success of such formulations to carve out a direction for this niche area to be explored further.
Collapse
Affiliation(s)
- Rajesh Kumar
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara - 144411, Punjab, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara - 144411, Punjab, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara - 144411, Punjab, India
| | - Deepika Sharma
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara - 144411, Punjab, India
| | - Omji Porwal
- Faculty of Pharmacy, Ishik University, Erbil, Iraq
| |
Collapse
|
20
|
Kohrt BA, Ottman K, Panter-Brick C, Konner M, Patel V. Why we heal: The evolution of psychological healing and implications for global mental health. Clin Psychol Rev 2020; 82:101920. [PMID: 33126037 DOI: 10.1016/j.cpr.2020.101920] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 09/02/2020] [Accepted: 09/13/2020] [Indexed: 01/10/2023]
Abstract
Why do humans heal one another? Evolutionary psychology has advanced our understanding of why humans suffer psychological distress and mental illness. However, to date, the evolutionary origins of what drives humans to alleviate the suffering of others has received limited attention. Therefore, we draw upon evolutionary theory to assess why humans psychologically support one another, focusing on the interpersonal regulation of emotions that shapes how humans heal and console one another when in psychosocial distress. To understand why we engage in psychological healing, we review the evolution of cooperation among social species and the roles of emotional contagion, empathy, and self-regulation. We discuss key aspects of human biocultural evolution that have contributed to healing behaviors: symbolic logic including language, complex social networks, and the long period of childhood that necessitates identifying and responding to others in distress. However, both biological and cultural evolution also have led to social context when empathy and consoling are impeded. Ultimately, by understanding the evolutionary processes shaping why humans psychologically do or do not heal one another, we can improve our current approaches in global mental health and uncover new opportunities to improve the treatment of mental illness across cultures and context around the world.
Collapse
Affiliation(s)
- Brandon A Kohrt
- Division of Global Mental Health, Department of Psychiatry and Behavioral Sciences, George Washington University, Washington, DC, USA.
| | - Katherine Ottman
- Division of Global Mental Health, Department of Psychiatry and Behavioral Sciences, George Washington University, Washington, DC, USA
| | - Catherine Panter-Brick
- Jackson Institute of Global Affairs, Yale University, New Haven, and Department of Anthropology, Yale University, New Haven, USA
| | - Melvin Konner
- Department of Anthropology, Emory University, Atlanta, USA
| | - Vikram Patel
- Department of Global Health and Social Medicine, Harvard Medical School, Boston, and Department of Global Health and Population, Harvard T. H. Chan School of Public Health, Harvard University, Boston, USA
| |
Collapse
|
21
|
Azhari A, Lim M, Bizzego A, Gabrieli G, Bornstein MH, Esposito G. Physical presence of spouse enhances brain-to-brain synchrony in co-parenting couples. Sci Rep 2020; 10:7569. [PMID: 32371912 PMCID: PMC7200679 DOI: 10.1038/s41598-020-63596-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 04/02/2020] [Indexed: 01/08/2023] Open
Abstract
Co-parenting spouses who live together remain in close physical proximity to each other and regularly engage in reciprocal social interactions in joint endeavors to coordinate their caregiving. Although bi-parental rearing is a common occurrence in humans, the influence of the physical presence of a co-parenting spouse on parental brain responses remains largely unknown. Synchrony is conceptualized as the matching of behavioral and physiological signals between two individuals. In this study, we examined how the presence of a co-parenting spouse influences brain-to-brain synchrony when attending to salient infant and adult vocalizations. We hypothesized that brain-to-brain synchrony would be greater in the presence of a spousal partner. Functional Near-infrared Spectroscopy (fNIRS) was used on 24 mother-father dyads (N = 48) to measure prefrontal cortical (PFC) activities while they listened to infant and adult vocalizations in two conditions, together (in the same room at the same time) and separately (in different rooms at different times). Couples showed greater synchrony in the together condition; when comparing fNIRS data between true couples and randomly matched controls, this synchronous effect was only seen in true couples, indicating a unique effect of spousal co-regulation toward salient stimuli. Our results indicate that the physical presence of the spouse might establish synchrony in attentional regulation mechanisms toward socially relevant stimuli. This finding holds implications for the role of the co-parenting spouse in influencing social and parental brain mechanisms.
Collapse
Affiliation(s)
- Atiqah Azhari
- Psychology Program, School of Social Sciences, Nanyang Technological University, Singapore, Singapore
| | - Mengyu Lim
- Psychology Program, School of Social Sciences, Nanyang Technological University, Singapore, Singapore
| | - Andrea Bizzego
- Division of Psychology, Department of Psychology and Cognitive Science, University of Trento, Trento, Italy
| | - Giulio Gabrieli
- Psychology Program, School of Social Sciences, Nanyang Technological University, Singapore, Singapore
| | - Marc H Bornstein
- National Institute of Child Health and Human Development, Bethesda, USA
- Institute for Fiscal Studies, London, United Kingdom
- UNICEF, New York, USA
| | - Gianluca Esposito
- Psychology Program, School of Social Sciences, Nanyang Technological University, Singapore, Singapore.
- Division of Psychology, Department of Psychology and Cognitive Science, University of Trento, Trento, Italy.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
22
|
Dolan SB, Berry MS, Johnson PS, Johnson MW. Potential for limited reinforcing and abuse-related subjective effects of intranasal oxytocin. J Psychopharmacol 2020; 34:336-347. [PMID: 31475622 PMCID: PMC8977072 DOI: 10.1177/0269881119867607] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND There has been growing interest in using oxytocin as a pharmacotherapy for psychiatric disorders, including substance use disorder. Limited data exist regarding oxytocin's reinforcing efficacy, which is a necessary consideration for novel pharmacotherapies, especially in substance-using populations. AIMS This study aimed to determine the potential reinforcing effects of intranasally administered oxytocin by assessing behavioral economic demand and subjective effects. METHODS Healthy adults (n = 23) participated in a double-blind, repeated-measures, laboratory study wherein they received intranasal oxytocin (40 IU) or placebo in a randomized order across two sessions. Participants completed drug purchasing tasks at the conclusion of both sessions. Throughout both sessions, subjective and physiological effects were assessed. RESULTS Demand-curve analysis of purchasing tasks revealed greater median purchasing for oxytocin relative to placebo. Physiological and subjective effects did not significantly differ between oxytocin and placebo. However, a nonsignificant trend was observed for moderately greater drug liking for oxytocin relative to placebo. There was a significant, positive correlation between the difference in drug liking (between oxytocin and placebo) and the difference in lowest-price purchasing (between oxytocin and placebo). CONCLUSIONS These data suggest the potential for limited reinforcing and abuse-related subjective effects of intranasal oxytocin. Given the small sample, the greater drug liking of oxytocin compared to placebo, and the positive relation between demand and drug liking, it is possible that oxytocin may produce reinforcing effects in some participants. Therefore, additional studies of oxytocin reinforcement are warranted.
Collapse
Affiliation(s)
- Sean B Dolan
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Meredith S Berry
- Department of Health Education and Behavior, University of Florida, Gainesville, FL, USA
- Department of Psychology, University of Florida, Gainesville, FL, USA
| | - Patrick S Johnson
- Department of Psychology, California State University, Chico, CA, USA
| | - Matthew W Johnson
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
23
|
Zhao W, Luo R, Sindermann C, Li J, Wei Z, Zhang Y, Liu C, Le J, Quintana DS, Montag C, Becker B, Kendrick KM. Oxytocin modulation of self-referential processing is partly replicable and sensitive to oxytocin receptor genotype. Prog Neuropsychopharmacol Biol Psychiatry 2020; 96:109734. [PMID: 31415827 DOI: 10.1016/j.pnpbp.2019.109734] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/18/2019] [Accepted: 08/10/2019] [Indexed: 11/25/2022]
Abstract
Intranasal oxytocin (OXT) has been associated with effects on diverse social-emotional domains in humans, however progress towards a therapeutic application of OXT in disorders with social-emotion impairments is currently hampered by poor replicability. Limited statistical power and individual differences in biological factors, such as oxytocin receptor (OXTR) genetics, may have contributed to these variable findings. To this end, employing a validated oxytocin-sensitive trait judgment paradigm, we present a pharmaco-genetic study aiming at (1) replicating previous findings suggesting that intranasal oxytocin (24 IU) reduces the self-referential bias in a large sample of n = 170 male subjects, (2) determining whether variations in common receptor polymorphisms (rs237887, rs2268491, rs2254298, rs53576, rs2268498) influence sensitivity to oxytocin's behavioral effects. We confirmed that in the whole sample oxytocin influenced self-other distinction in terms of reduced decision time. However, oxytocin only influenced decision time in rs53576 G carriers, whereas effects on subsequent memory performance were only found in rs2268498 TT homozygotes. In summary, the current study partially replicates our previous findings showing that oxytocin reduces the self-referential bias and suggests that sensitivity to its effects in this domain are receptor genotype dependent.
Collapse
Affiliation(s)
- Weihua Zhao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Ruixue Luo
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Cornelia Sindermann
- Department of Molecular Psychology, Institute of Psychology and Education, Ulm University, Helmholtzstr. 8/1, 89081 Ulm, Germany
| | - Jialin Li
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Zhenyu Wei
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Yingying Zhang
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Congcong Liu
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Jiao Le
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Daniel S Quintana
- NORMENT, KG Jebsen Centre for Psychosis Research, Division of Mental Health and Addiction, University of Oslo, and Oslo University Hospital, Oslo, Norway
| | - Christian Montag
- Department of Molecular Psychology, Institute of Psychology and Education, Ulm University, Helmholtzstr. 8/1, 89081 Ulm, Germany
| | - Benjamin Becker
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 611731, China.
| | - Keith M Kendrick
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, University of Electronic Science and Technology of China, Chengdu 611731, China.
| |
Collapse
|
24
|
Abstract
Abstract
Current psychological perspectives emphasize “attack” and “defense” as the behavioral mechanisms underlying conflict. Here, we extend this view by highlighting the relevance of pathological altruism and the neuroendocrine pathways associated with hostile behaviors. Specifically, we elucidate the modulatory role of the neuropeptide hormone oxytocin in motivating extraordinary levels of in-group commitment that can promote extreme behaviors and endure conflict with out-groups.
Collapse
|
25
|
Clinical potential of oxytocin in autism spectrum disorder: current issues and future perspectives. Behav Pharmacol 2019; 29:1-12. [PMID: 28857771 DOI: 10.1097/fbp.0000000000000341] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The effects of oxytocin on social cognition and behavior have recently attracted considerable attention. In particular, oxytocin has been proposed as a novel therapeutic for psychiatric disorders with social deficits such as autism spectrum disorders. This review provides a brief overview of behavioral and neural responses to oxytocin manipulations in humans and animal models. Although the differences in findings between human and animal studies should be interpreted carefully, shared behavioral phenotypes have been recognized, such as social bonding, social responses, and recognition and usage of social cues. Previous literature suggests that the neural effects of oxytocin in humans and animals overlap in the prefrontal, limbic, and paralimbic cortices. Oxytocin-induced alterations in these regions may indicate a fundamental basis for how oxytocin modulates social behaviors and facilitate the discovery of new pharmaceutical targets for treating social deficits.
Collapse
|
26
|
Seelke AM, Rhine MA, Khun K, Shweyk AN, Scott AM, Bond JM, Graham JL, Havel PJ, Wolden-Hanson T, Bales KL, Blevins JE. Intranasal oxytocin reduces weight gain in diet-induced obese prairie voles. Physiol Behav 2018; 196:67-77. [PMID: 30144467 DOI: 10.1016/j.physbeh.2018.08.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/17/2018] [Accepted: 08/19/2018] [Indexed: 12/17/2022]
Abstract
Oxytocin (OT) elicits weight loss in diet-induced obese (DIO) rodents, nonhuman primates and humans by reducing food intake and increasing energy expenditure. In addition to being important in the regulation of energy balance, OT is involved in social behaviors including parent-infant bonds, friendships, and pair bonds. However, the impact of social context on susceptibility to diet-induced obesity (DIO) and feeding behavior (including food sharing) has not been investigated in a rodent model that forms strong social bonds (i.e. prairie vole). Our goals were to determine in Prairie voles (Microtus ochrogaster) whether i) social context impacts susceptibility to DIO and ii) chronic intranasal OT reverses DIO. Voles were housed in divided cages with holes in the divider and paired with a same-sex animal with either the same food [high fat diet (HFD)/HFD, [low fat diet (LFD; chow)/chow], or the opposite food (HFD/chow or chow/HFD) for 19 weeks. HFD-fed voles pair-housed with voles maintained on the HFD demonstrated increased weight relative to pair-housed voles that were both maintained on chow. The study was repeated to determine the impact of social context on DIO susceptibility and body composition when animals are maintained on purified sugar-sweetened HFD and LFD to enhance palatability. As before, we found that voles demonstrated higher weight gain on the HFD/HFD housing paradigm, in part, through increased energy intake and the weight gain was a consequence of an increase in fat mass. However, HFD-fed animals housed with LFD-fed animals (and vice versa) showed intermediate patterns of weight gain and evidence of food sharing. Of translational importance is the finding that chronic intranasal OT appeared to reduce weight gain in DIO voles through a decrease in fat mass with no reduction in lean body mass. These effects were associated with transient reductions in food intake and increased food sharing. These findings identify a role of social context in the pathogenesis of DIO and indicate that chronic intranasal OT treatment reduces weight gain and body fat mass in DIO prairie voles, in part, by reducing food intake.
Collapse
Affiliation(s)
- Adele M Seelke
- Department of Psychology, University of California, Davis, CA, USA
| | - Maya A Rhine
- Department of Psychology, University of California, Davis, CA, USA
| | - Konterri Khun
- Department of Psychology, University of California, Davis, CA, USA
| | - Amira N Shweyk
- Department of Psychology, University of California, Davis, CA, USA
| | | | - Jessica M Bond
- Department of Psychology, University of California, Davis, CA, USA
| | - James L Graham
- Department of Nutrition and Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Peter J Havel
- Department of Nutrition and Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Tami Wolden-Hanson
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA
| | - Karen L Bales
- Department of Psychology, University of California, Davis, CA, USA
| | - James E Blevins
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
27
|
Freeman SM, Ngo J, Singh B, Masnaghetti M, Bales KL, Blevins JE. Effects of Chronic Oxytocin Administration and Diet Composition on Oxytocin and Vasopressin 1a Receptor Binding in the Rat Brain. Neuroscience 2018; 392:241-251. [PMID: 30071278 DOI: 10.1016/j.neuroscience.2018.07.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 07/19/2018] [Accepted: 07/20/2018] [Indexed: 12/19/2022]
Abstract
Oxytocin (OT) elicits weight loss in diet-induced obese (DIO) rodents, nonhuman primates, and humans, in part, by reducing food intake. Chronic OT administration produces more sustained weight loss in high-fat diet (HFD)-fed DIO rodents relative to chow-fed controls, but the reasons for this effect remain unclear. We hypothesized that HFD-induced obesity is associated with elevated OT receptor (OXTR) binding in brain regions where OT is known to cause decreased food intake and that this sensitized neural system is one mechanism by which OT preferentially elicits weight loss in DIO rodents. We therefore determined the impact of diet (HFD vs chow) and drug treatment (chronic OT infusion vs vehicle) on (1) OXTR binding in hindbrain and forebrain sites where OT suppresses food intake relative to control sites that express OXTR and (2) forebrain vasopressin 1a receptor (AVPR1a) density to evaluate the specificity of any OT effects. Using quantitative receptor autoradiography, we found that (1) diet composition failed to alter OXTR or AVPR1a binding; (2) chronic OT treatment produced largely global reductions in forebrain OXTR and AVPR1a binding without significantly altering hindbrain OXTR binding. These findings suggest that forebrain OXTR and AVPR1a are down-regulated in response to chronic OT treatment. Given that chronic intranasal OT may be used as a therapeutic strategy to treat obesity, future studies should consider the potential downregulatory effect that chronic treatment can have across forebrain and hindbrain nonapeptide receptors and assess the potential contribution of both receptor subtypes to the outcome measures.
Collapse
Affiliation(s)
- Sara M Freeman
- Department of Psychology, University of California, Davis, CA, USA
| | - Julie Ngo
- Department of Psychology, University of California, Davis, CA, USA
| | - Bhavdeep Singh
- Department of Psychology, University of California, Davis, CA, USA
| | | | - Karen L Bales
- Department of Psychology, University of California, Davis, CA, USA
| | - James E Blevins
- VA Puget Sound Health Care System, Office of Research and Development Medical Research Service, Department of Veterans Affairs Medical Center, Seattle, WA 98108, USA; Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
28
|
Tomaszycki ML, Atchley D. Pairing Increases Activation of V1aR, but not OTR, in Auditory Regions of Zebra Finches: The Importance of Signal Modality in Nonapeptide-Social Behavior Relationships. Integr Comp Biol 2018; 57:878-890. [PMID: 28992311 DOI: 10.1093/icb/icx043] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Social relationships are complex, involving the production and comprehension of signals, individual recognition, and close coordination of behavior between two or more individuals. The nonapeptides oxytocin and vasopressin are widely believed to regulate social relationships. These findings come largely from prairie voles, in which nonapeptide receptors in olfactory neural circuits drive pair bonding. This research is assumed to apply to all species. Previous reviews have offered two competing hypotheses. The work of Sarah Newman has implicated a common neural network across species, the Social Behavior Network. In contrast, others have suggested that there are signal modality-specific networks that regulate social behavior. Our research focuses on evaluating these two competing hypotheses in the zebra finch, a species that relies heavily on vocal/auditory signals for communication, specifically the neural circuits underlying singing in males and song perception in females. We have demonstrated that the quality of vocal interactions is highly important for the formation of long-term monogamous bonds in zebra finches. Qualitative evidence at first suggests that nonapeptide receptor distributions are very different between monogamous rodents (olfactory species) and monogamous birds (vocal/auditory species). However, we have demonstrated that social bonding behaviors are not only correlated with activation of nonapeptide receptors in vocal and auditory circuits, but also involve regions of the common Social Behavior Network. Here, we show increased Vasopressin 1a receptor, but not oxytocin receptor, activation in two auditory regions following formation of a pair bond. To our knowledge, this is the first study to suggest a role of nonapeptides in the auditory circuit in pair bonding. Thus, we highlight converging mechanisms of social relationships and also point to the importance of studying multiple species to understand mechanisms of behavior.
Collapse
Affiliation(s)
- Michelle L Tomaszycki
- Department of Psychology, Program in Neuroscience, Lafayette College, Easton, PA 18042, USA
| | - Derek Atchley
- Department of Psychology, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
29
|
Hurlemann R, Marsh N. Deciphering the modulatory role of oxytocin in human altruism. Rev Neurosci 2018; 28:335-342. [PMID: 28301323 DOI: 10.1515/revneuro-2016-0061] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Accepted: 01/06/2017] [Indexed: 11/15/2022]
Abstract
Unlike any other species, humans frequently engage in altruistic behaviors by which they increase another individual's welfare even if this implies personal costs. The psychological motives underlying altruistic behaviors remain diverse, ranging from the ability to reciprocate trust and cooperation to bonding and empathizing with family members or even genetically unrelated others. This article explores the neuroendocrine architecture of altruism by emphasizing the crucial role of the evolutionarily highly conserved peptide hormone oxytocin as a modulator of cooperative behaviors including empathy-driven altruism. However, accumulating evidence suggests that oxytocin does not invariably facilitate cooperation but also produces protective or even defensive-aggressive responses in specific social contexts. In addition, we highlight the relevance of message frames as critical determinants of whether the peptide promotes altruism toward prosocial ends.
Collapse
|
30
|
Jones C, Barrera I, Brothers S, Ring R, Wahlestedt C. Oxytocin and social functioning. DIALOGUES IN CLINICAL NEUROSCIENCE 2018. [PMID: 28867943 PMCID: PMC5573563 DOI: 10.31887/dcns.2017.19.2/cjones] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Social anxiety is a form of anxiety characterized by continuous fear of one or more social or performance situations. Although multiple treatment modalities (cognitive behavioral therapy, selective serotonin reuptake inhibitors/selective norepinephrine reuptake inhibitors, benzodiazepines) exist for social anxiety, they are effective for only 60% to 70% of patients. Thus, researchers have looked for other candidates for social anxiety treatment. Our review focuses on the peptide oxytocin as a potential therapeutic option for individuals with social anxiety. Animal research both in nonprimates and primates supports oxytocin's role in facilitation of prosocial behaviors and its anxiolytic effects. Human studies indicate significant associations between social anxiety and oxytocin receptor gene alleles, as well as social anxiety and oxytocin plasma levels. In addition, intranasal administration of oxytocin in humans has favorable effects on social anxiety symptomology. Other disorders, including autism, schizophrenia, and anorexia, have components of social anxiety in their pathophysiology. The therapeutic role of oxytocin for social dysfunction in these disorders is discussed.
Collapse
Affiliation(s)
- Candace Jones
- University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ingrid Barrera
- University of Miami Department of Psychiatry and Behavioral Sciences, Miami, Florida, USA
| | - Shaun Brothers
- University of Miami Department of Psychiatry and Behavioral Sciences, Miami, Florida, USA
| | - Robert Ring
- Drexel University Department of Pharmacology and Physiology, Philadelphia, Pennsylvania, USA
| | - Claes Wahlestedt
- University of Miami Department of Psychiatry and Behavioral Sciences, Miami, Florida, USA
| |
Collapse
|
31
|
Bowen MT, Neumann ID. Rebalancing the Addicted Brain: Oxytocin Interference with the Neural Substrates of Addiction. Trends Neurosci 2017; 40:691-708. [PMID: 29128108 DOI: 10.1016/j.tins.2017.10.003] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/05/2017] [Accepted: 10/06/2017] [Indexed: 12/21/2022]
Abstract
Drugs that act on the brain oxytocin (OXT) system may provide a much-needed treatment breakthrough for substance-use disorders. Targeting the brain OXT system has the potential to treat addiction to all major classes of addictive substance and to intervene across all stages of the addiction cycle. Emerging evidence suggests that OXT is able to interfere with such a wide range of addictive behaviours for such a wide range of addictive substances by rebalancing core neural systems that become dysregulated over the course of addiction. By improving our understanding of these interactions between OXT and the neural substrates of addiction, we will not only improve our understanding of addiction, but also our ability to effectively treat these devastating disorders.
Collapse
Affiliation(s)
- Michael T Bowen
- The University of Sydney, Faculty of Science, School of Psychology, Sydney, NSW, Australia; The University of Sydney, Brain and Mind Centre, Sydney, NSW, Australia
| | - Inga D Neumann
- Regensburg Center of Neuroscience, Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
32
|
Abstract
The oxytocin system has recently received increasing attention due to its effect on complex human behaviours. In parallel to this, over the past couple of decades, the human-analogue social behaviour of dogs has been intensively studied. Combining these two lines of research (e.g. studying the relationship between dog social behaviour and the oxytocin system) is a promising new research area. The present paper reviews the existing literature on how oxytocin is related to different aspects of human-directed social behaviour in dogs.
Collapse
Affiliation(s)
- Anna Kis
- Institute of Cognitive Neuroscience and Psychology, Hungarian Academy of Sciences, Magyar tudósok krt. 2, Budapest H-1117, Hungary.
| | - Alin Ciobica
- Department of Research, Faculty of Biology, Alexandru Ioan Cuza University, 11 Carol I Blvd., 700506, Iasi, Romania
| | - József Topál
- Institute of Cognitive Neuroscience and Psychology, Hungarian Academy of Sciences, Magyar tudósok krt. 2, Budapest H-1117, Hungary
| |
Collapse
|
33
|
Kamp-Becker I, Poustka L, Bachmann C, Ehrlich S, Hoffmann F, Kanske P, Kirsch P, Krach S, Paulus FM, Rietschel M, Roepke S, Roessner V, Schad-Hansjosten T, Singer T, Stroth S, Witt S, Wermter AK. Study protocol of the ASD-Net, the German research consortium for the study of Autism Spectrum Disorder across the lifespan: from a better etiological understanding, through valid diagnosis, to more effective health care. BMC Psychiatry 2017; 17:206. [PMID: 28577550 PMCID: PMC5455122 DOI: 10.1186/s12888-017-1362-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 05/19/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Autism Spectrum Disorder (ASD) is a severe, lifelong neurodevelopmental disorder with early onset that places a heavy burden on affected individuals and their families. Due to the need for highly specialized health, educational and vocational services, ASD is a cost-intensive disorder, and strain on health care systems increases with increasing age of the affected individual. METHODS The ASD-Net will study Germany's largest cohort of patients with ASD over the lifespan. By combining methodological expertise from all levels of clinical research, the ASD-Net will follow a translational approach necessary to identify neurobiological pathways of different phenotypes and their appropriate identification and treatment. The work of the ASD-Net will be organized into three clusters concentrating on diagnostics, therapy and health economics. In the diagnostic cluster, data from a large, well-characterized sample (N = 2568) will be analyzed to improve the efficiency of diagnostic procedures. Pattern classification methods (machine learning) will be used to identify algorithms for screening purposes. In a second step, the developed algorithm will be tested in an independent sample. In the therapy cluster, we will unravel how an ASD-specific social skills training with concomitant oxytocin administration can modulate behavior through neurobiological pathways. For the first time, we will characterize long-term effects of a social skills training combined with oxytocin treatment on behavioral and neurobiological phenotypes. Also acute effects of oxytocin will be investigated to delineate general and specific effects of additional oxytocin treatment in order to develop biologically plausible models for symptoms and successful therapeutic interventions in ASD. Finally, in the health economics cluster, we will assess service utilization and ASD-related costs in order to identify potential needs and cost savings specifically tailored to Germany. The ASD-Net has been established as part of the German Research Network for Mental Disorders, funded by the BMBF (German Federal Ministry of Education and Research). DISCUSSION The highly integrated structure of the ASD-Net guarantees sustained collaboration of clinicians and researchers to alleviate individual distress, harm, and social disability of patients with ASD and reduce costs to the German health care system. TRIAL REGISTRATION Both clinical trials of the ASD-Net are registered in the German Clinical Trials Register: DRKS00008952 (registered on August 4, 2015) and DRKS00010053 (registered on April 8, 2016).
Collapse
Affiliation(s)
- Inge Kamp-Becker
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Medical Clinic, Philipps-University Marburg, Marburg, Germany
| | - Luise Poustka
- Department of Child and Adolescent Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
- Department of Child and Adolescent Psychiatry, Medical University of Vienna, Vienna, Austria
- Department of Child and Adolescent Psychiatry/Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | | | - Stefan Ehrlich
- Department of Child & Adolescent Psychiatry, Medical Faculty of the Technical University Dresden, Dresden, Germany
- Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, TU Dresden, Dresden, Germany
| | - Falk Hoffmann
- Department of Health Services Research, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Philipp Kanske
- Department of Social Neuroscience, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Peter Kirsch
- Department of Clinical Psychology Central Institute of Mental Health, Mannheim, Germany
| | - Sören Krach
- Department for Psychiatry and Psychotherapy, University Schleswig-Holstein Campus Lübeck, Lübeck, Germany
| | - Frieder Michel Paulus
- Department for Psychiatry and Psychotherapy, University Schleswig-Holstein Campus Lübeck, Lübeck, Germany
| | - Marcella Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Mannheim, Germany
| | - Stefan Roepke
- Department of Psychiatry, Campus Benjamin Franklin, Charité - Medical Faculty Berlin, Berlin, Germany
| | - Veit Roessner
- Department of Child & Adolescent Psychiatry, Medical Faculty of the Technical University Dresden, Dresden, Germany
| | - Tanja Schad-Hansjosten
- Department of Child and Adolescent Psychiatry and Psychotherapy, Medical Faculty Mannheim, Central Institute of Mental Health, Heidelberg University, Mannheim, Germany
| | - Tania Singer
- Department of Social Neuroscience, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Sanna Stroth
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Medical Clinic, Philipps-University Marburg, Marburg, Germany
| | - Stephanie Witt
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Mannheim, Germany
| | - Anne-Kathrin Wermter
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Medical Clinic, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
34
|
Cherepanov SM, Yokoyama S, Mizuno A, Ichinose W, Lopatina O, Shabalova AA, Salmina AB, Yamamoto Y, Okamoto H, Shuto S, Higashida H. Structure-specific effects of lipidated oxytocin analogs on intracellular calcium levels, parental behavior, and oxytocin concentrations in the plasma and cerebrospinal fluid in mice. Pharmacol Res Perspect 2017; 5:e00290. [PMID: 28596839 PMCID: PMC5461640 DOI: 10.1002/prp2.290] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 11/25/2016] [Accepted: 11/29/2016] [Indexed: 12/27/2022] Open
Abstract
Oxytocin (OT) is a neuroendocrine nonapeptide that plays an important role in social memory and behavior. Nasal administration of OT has been shown to improve trust in healthy humans and social interaction in autistic subjects in some clinical trials. As a central nervous system (CNS) drug, however, OT has two unfavorable characteristics: OT is short‐acting and shows poor permeability across the blood–brain barrier, because it exists in charged form in the plasma and has short half‐life. To overcome these drawbacks, an analog with long‐lasting effects is required. We previously synthesized the analog, lipo‐oxytocin‐1 (LOT‐1), in which two palmitoyl groups are conjugated to the cysteine and tyrosine residues. In this study, we synthesized and evaluated the analogs lipo‐oxytocin‐2 (LOT‐2) and lipo‐oxytocin‐3 (LOT‐3), which feature the conjugation of one palmitoyl group at the cysteine and tyrosine residues, respectively. In human embryonic kidney‐293 cells overexpressing human OT receptors, these three LOTs demonstrated comparably weak effects on the elevation of intracellular free calcium concentrations after OT receptor activation, compared to the effects of OT. The three LOTs and OT exhibited different time‐dependent effects on recovery from impaired pup retrieval behavior in sires of CD38‐knockout mice. Sires treated with LOT‐1 showed the strongest effect, whereas others had no or little effects at 24 h after injection. These results indicated that LOTs have structure‐specific agonistic effects, and suggest that lipidation of OT might have therapeutic benefits for social impairment.
Collapse
Affiliation(s)
- Stanislav M Cherepanov
- Department of Basic Research on Social Recognition Research Center for Child Mental Development Kanazawa University Kanazawa 920-8640 Japan
| | - Shigeru Yokoyama
- Department of Basic Research on Social Recognition Research Center for Child Mental Development Kanazawa University Kanazawa 920-8640 Japan
| | - Akira Mizuno
- Faculty of Pharmaceutical Sciences Hokkaido University Kita-12, Nishi-6, Kita-ku Sapporo 060-0812 Japan
| | - Wataru Ichinose
- Faculty of Pharmaceutical Sciences Hokkaido University Kita-12, Nishi-6, Kita-ku Sapporo 060-0812 Japan
| | - Olga Lopatina
- Research Institute of Molecular Medicine & Pathobiochemistry Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky Krasnoyarsk 660022 Russia
| | - Anna A Shabalova
- Department of Basic Research on Social Recognition Research Center for Child Mental Development Kanazawa University Kanazawa 920-8640 Japan
| | - Alla B Salmina
- Department of Basic Research on Social Recognition Research Center for Child Mental Development Kanazawa University Kanazawa 920-8640 Japan.,Research Institute of Molecular Medicine & Pathobiochemistry Krasnoyarsk State Medical University named after Prof. V.F. Voino-Yasenetsky Krasnoyarsk 660022 Russia
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular BiologyGraduate School of Medical Sciences Kanazawa University Kanazawa 920-8640 Japan
| | - Hiroshi Okamoto
- Department of Biochemistry and Molecular Vascular BiologyGraduate School of Medical Sciences Kanazawa University Kanazawa 920-8640 Japan.,Department of Advanced Biological Sciences for Regeneration (Kotobiken Medical Laboratories) Tohoku University Graduate School of Medicine Sendai 980-8575 Japan
| | - Satoshi Shuto
- Faculty of Pharmaceutical Sciences Hokkaido University Kita-12, Nishi-6, Kita-ku Sapporo 060-0812 Japan.,Center for Research and Education on Drug Discovery Hokkaido University Kita-12, Nishi-6, Kita-ku Sapporo060-0812 Japan
| | - Haruhiro Higashida
- Department of Basic Research on Social Recognition Research Center for Child Mental Development Kanazawa University Kanazawa 920-8640 Japan
| |
Collapse
|
35
|
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder whose core symptoms include deficits in social interaction and communication besides restricted and repetitive behaviors. Although ASD is highly prevalent, affecting 1/100 in the general population, no medication for the core symptoms has been established. Therefore, the disorder is considered a huge unmet medical need and a heavy burden on individuals with ASD, their families, and entire society. Oxytocin is expected to be a potential therapeutic resource for the social core symptoms of ASD, since this neuropeptide can modulate human social behavior and cognition. This review article provides an overview of both experimental and clinical studies on effects of oxytocin administration on behavior, neural underpinnings, and symptomatology of ASD. Although the number of studies is increasing, several issues remain for further development of clinical application of the neuropeptide. The issues include optimization of administration route, doses, treatment duration, interval of administrations, and timing of starting treatment. Additional issues involve investigating neurobiological mechanisms of treatment and developing a reliable tool to accurately and objectively assess longitudinal changes in the core symptoms of ASD. Some of these issues are discussed in this review.
Collapse
Affiliation(s)
- Hidenori Yamasue
- Department of Psychiatry, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu, Shizuoka, 431-3192, Japan.
| | - Gregor Domes
- Department of Biological and Clinical Psychology, University of Trier, Am Johanniterufer 15, 54290, Trier, Germany.
| |
Collapse
|
36
|
Abstract
Autism spectrum disorders (ASDs) are neurodevelopmental disorders with early onset, characterized by deficits in social communication and repetitive and restricted interests and activities. A growing number of studies over the last 10 years support the efficacy of behaviorally based interventions in ASD for the improvement of social communication and behavioral functioning. In contrast, research on neurobiological based therapies for ASD is still at its beginnings. In this article, we will provide a selective overview of both well-established evidence-based treatments and novel interventions and drug treatments based on neurobiological principles aiming at improving core symptoms in ASD. Directions and options for future research on treatment in ASD are discussed.
Collapse
Affiliation(s)
- L Poustka
- Clinic for Child and Adolescent Psychiatry, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| | - I Kamp-Becker
- Clinic for Child and Adolescent Psychiatry, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| |
Collapse
|
37
|
Sasaki T, Hashimoto K, Oda Y, Ishima T, Yakita M, Kurata T, Kunou M, Takahashi J, Kamata Y, Kimura A, Niitsu T, Komatsu H, Hasegawa T, Shiina A, Hashimoto T, Kanahara N, Shimizu E, Iyo M. Increased Serum Levels of Oxytocin in 'Treatment Resistant Depression in Adolescents (TRDIA)' Group. PLoS One 2016; 11:e0160767. [PMID: 27536785 PMCID: PMC4990411 DOI: 10.1371/journal.pone.0160767] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Accepted: 07/25/2016] [Indexed: 11/18/2022] Open
Abstract
Objective ‘Treatment-resistant depression’ is depression that does not respond to an adequate regimen of evidence-based treatment. Treatment-resistant depression frequently becomes chronic. Children with treatment-resistant depression might also develop bipolar disorder (BD). The objective of this study was to determine whether serum levels of oxytocin (OXT) in treatment-resistant depression in adolescents (TRDIA) differ from non-treatment-resistant depression in adolescents (non-TRDIA) or controls. We also investigated the relationships between serum OXT levels and the clinical symptoms, severity, and familial histories of adolescent depressive patients. Methods We measured serum OXT levels: TRDIA (n = 10), non-TRDIA (n = 27), and age- and sex- matched, neurotypical controls (n = 25). Patients were evaluated using the Children’s Depression Rating Scale-Revised (CDRS-R) and the Depression Self-Rating Scale for Children-Japanese Version (DSRS-C-J). The patients were also assessed retrospectively using the following variables: familial history of major depressive disorder and BD (1st degree or 2nd degree), history of disruptive mood dysregulation disorder, recurrent depressive disorder (RDD), history of antidepressant activation. Results Serum levels of OXT among the TRDIA and non-TRDIA patients and controls differed significantly. Interestingly, the rates of a family history of BD (1st or 2nd degree), RDD and a history of antidepressant activation in our TRDIA group were significantly higher than those of the non-TRDIA group. Conclusions Serum levels of OXT may play a role in the pathophysiology of TRDIA.
Collapse
Affiliation(s)
- Tsuyoshi Sasaki
- Department of Child Psychiatry, Chiba University Hospital, Chiba, Japan
- Departments of Psychiatry Chiba University Graduate School of Medicine, Chiba, Japan
- * E-mail:
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Yasunori Oda
- Departments of Psychiatry Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tamaki Ishima
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Madoka Yakita
- Department of Child Psychiatry, Chiba University Hospital, Chiba, Japan
| | - Tsutomu Kurata
- Department of Child Psychiatry, Chiba University Hospital, Chiba, Japan
| | - Masaru Kunou
- Department of Child Psychiatry, Chiba University Hospital, Chiba, Japan
- Departments of Cognitive Behavioral Physiology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Jumpei Takahashi
- Department of Child Psychiatry, Chiba University Hospital, Chiba, Japan
| | - Yu Kamata
- Departments of Psychiatry Chiba University Graduate School of Medicine, Chiba, Japan
| | - Atsushi Kimura
- Departments of Psychiatry Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tomihisa Niitsu
- Departments of Psychiatry Chiba University Graduate School of Medicine, Chiba, Japan
| | - Hideki Komatsu
- Departments of Psychiatry Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tadashi Hasegawa
- Departments of Psychiatry Chiba University Graduate School of Medicine, Chiba, Japan
| | - Akihiro Shiina
- Departments of Psychiatry Chiba University Graduate School of Medicine, Chiba, Japan
| | - Tasuku Hashimoto
- Departments of Psychiatry Chiba University Graduate School of Medicine, Chiba, Japan
| | - Nobuhisa Kanahara
- Division of Medical Treatment and Rehabilitation, Chiba University Center for Forensic Mental Health, Chiba, Japan
| | - Eiji Shimizu
- Departments of Cognitive Behavioral Physiology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Masaomi Iyo
- Department of Child Psychiatry, Chiba University Hospital, Chiba, Japan
- Departments of Psychiatry Chiba University Graduate School of Medicine, Chiba, Japan
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba, Japan
- Division of Medical Treatment and Rehabilitation, Chiba University Center for Forensic Mental Health, Chiba, Japan
| |
Collapse
|
38
|
Kosaka H, Okamoto Y, Munesue T, Yamasue H, Inohara K, Fujioka T, Anme T, Orisaka M, Ishitobi M, Jung M, Fujisawa TX, Tanaka S, Arai S, Asano M, Saito DN, Sadato N, Tomoda A, Omori M, Sato M, Okazawa H, Higashida H, Wada Y. Oxytocin efficacy is modulated by dosage and oxytocin receptor genotype in young adults with high-functioning autism: a 24-week randomized clinical trial. Transl Psychiatry 2016; 6:e872. [PMID: 27552585 PMCID: PMC5022092 DOI: 10.1038/tp.2016.152] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 06/22/2016] [Accepted: 07/04/2016] [Indexed: 12/27/2022] Open
Abstract
Recent studies have suggested that long-term oxytocin administration can alleviate the symptoms of autism spectrum disorder (ASD); however, factors influencing its efficacy are still unclear. We conducted a single-center phase 2, pilot, randomized, double-blind, placebo-controlled, parallel-group, clinical trial in young adults with high-functioning ASD, to determine whether oxytocin dosage and genetic background of the oxytocin receptor affects oxytocin efficacy. This trial consisted of double-blind (12 weeks), open-label (12 weeks) and follow-up phases (8 weeks). To examine dose dependency, 60 participants were randomly assigned to high-dose (32 IU per day) or low-dose intranasal oxytocin (16 IU per day), or placebo groups during the double-blind phase. Next, we measured single-nucleotide polymorphisms (SNPs) in the oxytocin receptor gene (OXTR). In the intention-to-treat population, no outcomes were improved after oxytocin administration. However, in male participants, Clinical Global Impression-Improvement (CGI-I) scores in the high-dose group, but not the low-dose group, were significantly higher than in the placebo group. Furthermore, we examined whether oxytocin efficacy, reflected in the CGI-I scores, is influenced by estimated daily dosage and OXTR polymorphisms in male participants. We found that >21 IU per day oxytocin was more effective than ⩽21 IU per day, and that a SNP in OXTR (rs6791619) predicted CGI-I scores for ⩽21 IU per day oxytocin treatment. No severe adverse events occurred. These results suggest that efficacy of long-term oxytocin administration in young men with high-functioning ASD depends on the oxytocin dosage and genetic background of the oxytocin receptor, which contributes to the effectiveness of oxytocin treatment of ASD.
Collapse
Affiliation(s)
- H Kosaka
- Research Center for Child Mental Development, University of Fukui, Eiheiji, Japan,Department of Neuropsychiatry, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan,Division of Developmental Higher Brain Functions, Department of Child Development United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Eiheiji, Japan,Research Center for Child Mental Development, University of Fukui, 23-3 Matsuokashimoaizuki, Eiheiji 910-1193, Fukui, Japan. E-mail:
| | - Y Okamoto
- Research Center for Child Mental Development, University of Fukui, Eiheiji, Japan,Division of Developmental Higher Brain Functions, Department of Child Development United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Eiheiji, Japan
| | - T Munesue
- Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - H Yamasue
- Department of Neuropsychiatry, School of Medicine, University of Tokyo, Tokyo, Japan
| | - K Inohara
- Department of Neuropsychiatry, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan,Department of Informatics, Graduate School of Informatics and Engineering, The University of Electro-Communications, Chofu, Japan
| | - T Fujioka
- Department of Neuropsychiatry, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan
| | - T Anme
- International Community Care and Lifespan Development, Empowerment Sciences, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - M Orisaka
- Department of Obstetrics and Gynecology, University of Fukui, Eiheiji, Japan
| | - M Ishitobi
- Department of Neuropsychiatry, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan,Department of Child and Adolescent Mental Health, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - M Jung
- Research Center for Child Mental Development, University of Fukui, Eiheiji, Japan,Division of Developmental Higher Brain Functions, Department of Child Development United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Eiheiji, Japan
| | - T X Fujisawa
- Research Center for Child Mental Development, University of Fukui, Eiheiji, Japan,Division of Developmental Higher Brain Functions, Department of Child Development United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Eiheiji, Japan
| | - S Tanaka
- Research Center for Child Mental Development, University of Fukui, Eiheiji, Japan
| | - S Arai
- Research Center for Child Mental Development, University of Fukui, Eiheiji, Japan,Division of Developmental Higher Brain Functions, Department of Child Development United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Eiheiji, Japan
| | - M Asano
- Research Center for Child Mental Development, University of Fukui, Eiheiji, Japan,Department of Neuropsychiatry, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan,Division of Developmental Higher Brain Functions, Department of Child Development United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Eiheiji, Japan
| | - D N Saito
- Research Center for Child Mental Development, University of Fukui, Eiheiji, Japan,Division of Developmental Higher Brain Functions, Department of Child Development United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Eiheiji, Japan,Biomedical Imaging Research Center, University of Fukui, Eiheiji, Japan
| | - N Sadato
- Department of Cerebral Research, National Institute for Physiological Sciences, Okazaki, Japan
| | - A Tomoda
- Research Center for Child Mental Development, University of Fukui, Eiheiji, Japan,Division of Developmental Higher Brain Functions, Department of Child Development United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Eiheiji, Japan
| | - M Omori
- Faculty of Nursing and Social Welfare Sciences, Fukui Prefectural University, Eiheiji, Japan
| | - M Sato
- Research Center for Child Mental Development, University of Fukui, Eiheiji, Japan,Division of Developmental Higher Brain Functions, Department of Child Development United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Eiheiji, Japan,Department of Anatomy and Neuroscience, Graduate School of Medicine, Osaka University, Suita, Japan,Division of Developmental Neuroscience, Department of Child Development United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Suita, Japan
| | - H Okazawa
- Research Center for Child Mental Development, University of Fukui, Eiheiji, Japan,Division of Developmental Higher Brain Functions, Department of Child Development United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Eiheiji, Japan,Biomedical Imaging Research Center, University of Fukui, Eiheiji, Japan
| | - H Higashida
- Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan
| | - Y Wada
- Research Center for Child Mental Development, University of Fukui, Eiheiji, Japan,Department of Neuropsychiatry, Faculty of Medical Sciences, University of Fukui, Eiheiji, Japan,Division of Developmental Higher Brain Functions, Department of Child Development United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Eiheiji, Japan
| |
Collapse
|
39
|
Zhong J, Amina S, Liang M, Akther S, Yuhi T, Nishimura T, Tsuji C, Tsuji T, Liu HX, Hashii M, Furuhara K, Yokoyama S, Yamamoto Y, Okamoto H, Zhao YJ, Lee HC, Tominaga M, Lopatina O, Higashida H. Cyclic ADP-Ribose and Heat Regulate Oxytocin Release via CD38 and TRPM2 in the Hypothalamus during Social or Psychological Stress in Mice. Front Neurosci 2016; 10:304. [PMID: 27499729 PMCID: PMC4956647 DOI: 10.3389/fnins.2016.00304] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/16/2016] [Indexed: 12/20/2022] Open
Abstract
Hypothalamic oxytocin (OT) is released into the brain by cyclic ADP-ribose (cADPR) with or without depolarizing stimulation. Previously, we showed that the intracellular free calcium concentration ([Ca2+]i) that seems to trigger OT release can be elevated by β-NAD+, cADPR, and ADP in mouse oxytocinergic neurons. As these β-NAD+ metabolites activate warm-sensitive TRPM2 cation channels, when the incubation temperature is increased, the [Ca2+]i in hypothalamic neurons is elevated. However, it has not been determined whether OT release is facilitated by heat in vitro or hyperthermia in vivo in combination with cADPR. Furthermore, it has not been examined whether CD38 and TRPM2 exert their functions on OT release during stress or stress-induced hyperthermia in relation to the anxiolytic roles and social behaviors of OT under stress conditions. Here, we report that OT release from the isolated hypothalami of male mice in culture was enhanced by extracellular application of cADPR or increasing the incubation temperature from 35°C to 38.5°C, and simultaneous stimulation showed a greater effect. This release was inhibited by a cADPR-dependent ryanodine receptor inhibitor and a nonspecific TRPM2 inhibitor. The facilitated release by heat and cADPR was suppressed in the hypothalamus isolated from CD38 knockout mice and CD38- or TRPM2-knockdown mice. In the course of these experiments, we noted that OT release differed markedly between individual mice under stress with group housing. That is, when male mice received cage-switch stress and eliminated due to their social subclass, significantly higher levels of OT release were found in subordinates compared with ordinates. In mice exposed to anxiety stress in an open field, the cerebrospinal fluid (CSF) OT level increased transiently at 5 min after exposure, and the rectal temperature also increased from 36.6°C to 37.8°C. OT levels in the CSF of mice with lipopolysaccharide-induced fever (+0.8°C) were higher than those of control mice. The TRPM2 mRNA levels and immunoreactivities increased in the subordinate group with cage-switch stress. These results showed that cADPR/CD38 and heat/TRPM2 are co-regulators of OT secretion and suggested that CD38 and TRPM2 are potential therapeutic targets for OT release in psychiatric diseases caused by social stress.
Collapse
Affiliation(s)
- Jing Zhong
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Sarwat Amina
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Mingkun Liang
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Shirin Akther
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Teruko Yuhi
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Tomoko Nishimura
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Chiharu Tsuji
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Takahiro Tsuji
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Hong-Xiang Liu
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Minako Hashii
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Kazumi Furuhara
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Shigeru Yokoyama
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa University Kanazawa, Japan
| | - Yasuhiko Yamamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences Kanazawa, Japan
| | - Hiroshi Okamoto
- Department of Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical SciencesKanazawa, Japan; Department of Biochemistry, Tohoku University Graduate School of MedicineSendai, Japan
| | - Yong Juan Zhao
- School of Chemical Biology and Biotechnology, Peking University Graduate School Shenzhen, China
| | - Hon Cheung Lee
- School of Chemical Biology and Biotechnology, Peking University Graduate School Shenzhen, China
| | - Makoto Tominaga
- Division of Cell Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences Okazaki, Japan
| | - Olga Lopatina
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa UniversityKanazawa, Japan; Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical UniversityKrasnoyarsk, Russia
| | - Haruhiro Higashida
- Department of Basic Research on Social Recognition and Memory, Research Centre for Child Mental Development, Kanazawa UniversityKanazawa, Japan; Research Institute of Molecular Medicine and Pathobiochemistry, Krasnoyarsk State Medical UniversityKrasnoyarsk, Russia
| |
Collapse
|
40
|
Lee JH, Espinera AR, Chen D, Choi KE, Caslin AY, Won S, Pecoraro V, Xu GY, Wei L, Yu SP. Neonatal inflammatory pain and systemic inflammatory responses as possible environmental factors in the development of autism spectrum disorder of juvenile rats. J Neuroinflammation 2016; 13:109. [PMID: 27184741 PMCID: PMC4867541 DOI: 10.1186/s12974-016-0575-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/06/2016] [Indexed: 02/28/2023] Open
Abstract
Background Autism spectrum disorder (ASD) affects many children and juveniles. The pathogenesis of ASD is not well understood. Environmental factors may play important roles in the development of ASD. We examined a possible relationship of inflammatory pain in neonates and the development of ASD in juveniles. Methods Acute inflammation pain was induced by 5 % formalin (5 μl/day) subcutaneous injection into two hindpaws of postnatal day 3 to 5 (P3–P5) rat pups. Western blot, immunohistochemical, and behavioral examinations were performed at different time points after the insult. Results Formalin injection caused acute and chronic inflammatory responses including transient local edema, increased levels of inflammatory cytokines, TNF-α, and IL-1β in the blood as well as in the brain, and increased microglia in the brain. One day after the pain insult, there was significant cell death in the cortex and hippocampus. Two weeks later, although the hindpaw local reaction subsided, impaired axonal growth and demyelization were seen in the brain of P21 juvenile rats. The number of bromodeoxyuridine (BrdU) and doublecortin (DCX) double-positive cells in the hippocampal dentate gyrus of P21 rats was significantly lower than that in controls, indicating reduced neurogenesis. In the P21 rat’s brain of the formalin group, the expression of autism-related gene neurexin 1 (NRXN1), fragile X mental retardation 1 (FMR1), and oxytocin was significantly downregulated, consistent with the gene alteration in ASD. Juvenile rats in the formalin group showed hyperalgesia, repetitive behaviors, abnormal locomotion, sleep disorder, and distinct deficits in social memory and social activities. These alterations in neuroinflammatory reactions, gene expression, and behaviors were more evident in male than in female rats. Importantly, an anti-inflammation treatment using indomethacin (10 mg/kg, i.p.) at the time of formalin injections suppressed inflammatory responses and neuronal cell death and prevented alterations in ASD-related genes and the development of abnormal behaviors. Conclusions These novel observations indicate that severe inflammatory pain in neonates and persistent inflammatory reactions may predispose premature infants to development delays and psychiatric disorders including ASD. The prevention of pain stimuli and prompt treatments of inflammation during development appear vitally important in disrupting possible evolution of ASD syndromes. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0575-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jin Hwan Lee
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Alyssa R Espinera
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Dongdong Chen
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,The Laboratory of Translational Pain Medicine, Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Ko-Eun Choi
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Asha Yoshiko Caslin
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Soonmi Won
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Valentina Pecoraro
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Guang-Yin Xu
- The Laboratory of Translational Pain Medicine, Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Ling Wei
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, 30322, USA. .,Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Atlanta, GA, 30033, USA. .,Emory University School of Medicine, 101 Woodruff Circle, WMB Suite 620, Atlanta, GA, 30322, USA.
| |
Collapse
|
41
|
Aoki Y, Cortese S, Tansella M. Neural bases of atypical emotional face processing in autism: A meta-analysis of fMRI studies. World J Biol Psychiatry 2016; 16:291-300. [PMID: 25264291 DOI: 10.3109/15622975.2014.957719] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVES We aim to outline the neural correlates of atypical emotional face processing in individuals with ASD. METHODS A comprehensive literature search was conducted through electronic databases to identify functional magnetic resonance imaging (fMRI) studies of whole brain analysis with emotional-face processing tasks in individuals with ASD. The Signed Differential Mapping with random effects model was used to conduct meta-analyses. Identified fMRI studies were further divided into sub-groups based on contrast ("emotional-face vs. non-emotional-face" or "emotional-face vs. non-face") to confirm the results of a meta-analysis of the whole studies. RESULTS Thirteen studies with 226 individuals with ASD and 251 typically developing people were identified. We found ASD-related hyperactivation in subcortical structures, including bilateral thalamus, bilateral caudate, and right precuneus, and ASD-related hypoactivation in the hypothalamus during emotional-face processing. Sub-analyses with more homogeneous contrasts preserved the findings of the main analysis such as hyperactivation in sub-cortical structure. Jackknife analyses showed that hyperactivation of the left caudate was the most robust finding. CONCLUSIONS Abnormalities in the subcortical structures, such as amygdala, hypothalamus and basal ganglia, are associated with atypical emotional-face processing in individuals with ASD.
Collapse
Affiliation(s)
- Yuta Aoki
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo , Bunkyo-ku, Tokyo , Japan
| | | | | |
Collapse
|
42
|
Fuccillo MV. Striatal Circuits as a Common Node for Autism Pathophysiology. Front Neurosci 2016; 10:27. [PMID: 26903795 PMCID: PMC4746330 DOI: 10.3389/fnins.2016.00027] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 01/22/2016] [Indexed: 12/17/2022] Open
Abstract
Autism spectrum disorders (ASD) are characterized by two seemingly unrelated symptom domains-deficits in social interactions and restrictive, repetitive patterns of behavioral output. Whether the diverse nature of ASD symptomatology represents distributed dysfunction of brain networks or abnormalities within specific neural circuits is unclear. Striatal dysfunction is postulated to underlie the repetitive motor behaviors seen in ASD, and neurological and brain-imaging studies have supported this assumption. However, as our appreciation of striatal function expands to include regulation of behavioral flexibility, motivational state, goal-directed learning, and attention, we consider whether alterations in striatal physiology are a central node mediating a range of autism-associated behaviors, including social and cognitive deficits that are hallmarks of the disease. This review investigates multiple genetic mouse models of ASD to explore whether abnormalities in striatal circuits constitute a common pathophysiological mechanism in the development of autism-related behaviors. Despite the heterogeneity of genetic insult investigated, numerous genetic ASD models display alterations in the structure and function of striatal circuits, as well as abnormal behaviors including repetitive grooming, stereotypic motor routines, deficits in social interaction and decision-making. Comparative analysis in rodents provides a unique opportunity to leverage growing genetic association data to reveal canonical neural circuits whose dysfunction directly contributes to discrete aspects of ASD symptomatology. The description of such circuits could provide both organizing principles for understanding the complex genetic etiology of ASD as well as novel treatment routes. Furthermore, this focus on striatal mechanisms of behavioral regulation may also prove useful for exploring the pathogenesis of other neuropsychiatric diseases, which display overlapping behavioral deficits with ASD.
Collapse
Affiliation(s)
- Marc V. Fuccillo
- Department of Neuroscience, Perelman School of Medicine, University of PennsylvaniaPhiladelphia, PA, USA
| |
Collapse
|
43
|
Rothwell PE. Autism Spectrum Disorders and Drug Addiction: Common Pathways, Common Molecules, Distinct Disorders? Front Neurosci 2016; 10:20. [PMID: 26903789 PMCID: PMC4742554 DOI: 10.3389/fnins.2016.00020] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/15/2016] [Indexed: 11/17/2022] Open
Abstract
Autism spectrum disorders (ASDs) and drug addiction do not share substantial comorbidity or obvious similarities in etiology or symptomatology. It is thus surprising that a number of recent studies implicate overlapping neural circuits and molecular signaling pathways in both disorders. The purpose of this review is to highlight this emerging intersection and consider implications for understanding the pathophysiology of these seemingly distinct disorders. One area of overlap involves neural circuits and neuromodulatory systems in the striatum and basal ganglia, which play an established role in addiction and reward but are increasingly implicated in clinical and preclinical studies of ASDs. A second area of overlap relates to molecules like Fragile X mental retardation protein (FMRP) and methyl CpG-binding protein-2 (MECP2), which are best known for their contribution to the pathogenesis of syndromic ASDs, but have recently been shown to regulate behavioral and neurobiological responses to addictive drug exposure. These shared pathways and molecules point to common dimensions of behavioral dysfunction, including the repetition of behavioral patterns and aberrant reward processing. The synthesis of knowledge gained through parallel investigations of ASDs and addiction may inspire the design of new therapeutic interventions to correct common elements of striatal dysfunction.
Collapse
Affiliation(s)
- Patrick E Rothwell
- Department of Neuroscience, University of Minnesota Minneapolis, MN, USA
| |
Collapse
|
44
|
Yamasue H. Promising evidence and remaining issues regarding the clinical application of oxytocin in autism spectrum disorders. Psychiatry Clin Neurosci 2016; 70:89-99. [PMID: 26394796 DOI: 10.1111/pcn.12364] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/18/2015] [Indexed: 12/24/2022]
Abstract
Oxytocin is a potential therapeutic for the core symptoms of autism spectrum disorder (ASD), which is currently untreatable with pharmacotherapy. Previous clinical trials of a single dose of oxytocin have consistently reported significantly positive effects on various experimental measures associated with the core symptoms of ASD. These studies used various experimental measures as surrogate endpoints of the trials. However, to date, randomized clinical trials of continual administration of oxytocin have failed to reveal significant positive effects on clinically meaningful endpoints, such as how those with ASD interact during interpersonal interactions. This article reviews both the negative and positive effects of oxytocin on the core symptoms of ASD and their surrogate markers. Some unresolved and critical issues on the development of oxytocin as a new therapeutic have been extracted: optimization of dose, duration of oxytocin treatment, and the development of objective and reliable measurements of clinically meaningful endpoints for the core symptoms of ASD. Furthermore, optimization to the intranasal delivery system and careful consideration of how individuals respond differently to treatments should be addressed in future studies.
Collapse
Affiliation(s)
- Hidenori Yamasue
- Department of Neuropsychiatry, School of Medicine, The University of Tokyo, Tokyo, Japan.,Japan Science and Technology Agency, CREST, Tokyo, Japan
| |
Collapse
|
45
|
Munesue T, Nakamura H, Kikuchi M, Miura Y, Takeuchi N, Anme T, Nanba E, Adachi K, Tsubouchi K, Sai Y, Miyamoto KI, Horike SI, Yokoyama S, Nakatani H, Niida Y, Kosaka H, Minabe Y, Higashida H. Oxytocin for Male Subjects with Autism Spectrum Disorder and Comorbid Intellectual Disabilities: A Randomized Pilot Study. Front Psychiatry 2016; 7:2. [PMID: 26834651 PMCID: PMC4720778 DOI: 10.3389/fpsyt.2016.00002] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 01/05/2016] [Indexed: 12/27/2022] Open
Abstract
UNLABELLED Approximately half of autism spectrum disorder (ASD) individuals suffer from comorbid intellectual disabilities (IDs). Oxytocin (OXT) receptors are highly expressed in temporal lobe structures and are likely to play a modulatory role in excitatory/inhibitory balance, at least based on animal model findings. Thus, it is feasible that in the highly representative group of Kanner-type ASD subjects, OXT could have a beneficial effect on social communication and social interaction. The aim of this pilot study is to investigate the feasibility and adverse events, such as epilepsy, of the long-term administration of intranasal OXT for adolescent and adult ASD subjects with ID because such patients frequently have seizures. We also addressed the question on how to scale the OXT effects to the core symptoms of social deficits because of the relative difficulty in obtaining objective measurements. Twenty-nine males (aged 15-40 years old) participated in a randomized, double-blind, and placebo-controlled crossover study (each for 8 weeks) with OXT (16 IU/day). Except for seizures experienced by one participant, other serious adverse events did not occur. The primary and secondary outcomes measured using the Childhood Autism Rating Scale and several standard scales, respectively, revealed no difference between the OXT and placebo groups. Instead, in an exploratory analysis, the social interactions observed in the play sessions or in daily life were significantly more frequent in the initial half period in the OXT-first arm of the crossover trial. There were also significant correlations between the plasma OXT concentration and subscale scores for irritability on the Aberrant Behavior Checklist. In conclusion, this pilot study demonstrates that long-term administration of intranasal OXT is tolerable in a representative cohort of ASD individuals with ID and suggests that future multicenter trials of OXT are warranted and should include measurements of reciprocal social interactions based on daily life under closer surveillance for epilepsy. TRIAL REGISTRATION UMIN000007250.
Collapse
Affiliation(s)
- Toshio Munesue
- Research Center for Child Mental Development, Kanazawa University , Kanazawa , Japan
| | - Hiroyuki Nakamura
- Department of Environmental and Preventive Medicine, Graduate School of Medical Sciences, Kanazawa University , Kanazawa , Japan
| | - Mitsuru Kikuchi
- Research Center for Child Mental Development, Kanazawa University , Kanazawa , Japan
| | - Yui Miura
- Research Center for Child Mental Development, Kanazawa University , Kanazawa , Japan
| | - Noriyuki Takeuchi
- Research Center for Child Mental Development, Kanazawa University , Kanazawa , Japan
| | - Tokie Anme
- International Community Care and Lifespan Development, Empowerment Sciences, Faculty of Medicine, University of Tsukuba , Tsukuba , Japan
| | - Eiji Nanba
- Division of Functional Genomics, Research Center for Bioscience and Technology, Tottori University , Yonago , Japan
| | - Kaori Adachi
- Division of Functional Genomics, Research Center for Bioscience and Technology, Tottori University , Yonago , Japan
| | | | - Yoshimichi Sai
- Department of Pharmacy, Kanazawa University Hospital , Kanazawa , Japan
| | - Ken-Ichi Miyamoto
- Department of Pharmacy, Kanazawa University Hospital , Kanazawa , Japan
| | - Shin-Ichi Horike
- Advanced Science Research Center, Kanazawa University , Kanazawa , Japan
| | - Shigeru Yokoyama
- Research Center for Child Mental Development, Kanazawa University , Kanazawa , Japan
| | - Hideo Nakatani
- Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan; Department of Psychiatry and Neurobiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yo Niida
- Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan; Division of Genomic Medicine, Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| | - Hirotaka Kosaka
- Research Center for Child Mental Development, University of Fukui , Fukui , Japan
| | - Yoshio Minabe
- Research Center for Child Mental Development, Kanazawa University, Kanazawa, Japan; Department of Psychiatry and Neurobiology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Haruhiro Higashida
- Research Center for Child Mental Development, Kanazawa University , Kanazawa , Japan
| |
Collapse
|
46
|
Brüne M. On the role of oxytocin in borderline personality disorder. BRITISH JOURNAL OF CLINICAL PSYCHOLOGY 2015; 55:287-304. [PMID: 26616386 DOI: 10.1111/bjc.12100] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 10/21/2015] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Interpersonal dysfunction is central to borderline personality disorder (BPD). Recent research has focused on the role of oxytocin (OT) in BPD, particularly regarding associations of OT activity with symptoms, genetic polymorphisms of the oxytocin receptor coding gene (OXTR) in BPD, and experimental modification of interpersonal core problems of patients with BPD such as hypervigilance towards threat detection, mistrust, and non-verbal behaviour during social interaction by intranasal application of OT. METHODS A literature ('medline') review was performed using the keywords 'oxytocin' and 'borderline personality disorder'. Secondary literature on trauma and attachment in relation to OT was also considered relevant. RESULTS Together, findings suggest that in BPD OT is associated with enhanced defensive mechanisms and avoidance behaviour. Moreover, gene-environment interaction concerning polymorphic variations of the OXTR gene and childhood adversity in BPD suggests that these genes convey developmental flexibility or 'differential susceptibility' to environmental contingencies, whereby BPD resides at the poor outcome end of the spectrum. CONCLUSIONS In view of the conflicting literature, it needs to be studied carefully whether OT can serve as a therapeutic agent given adjunct to psychotherapy in BPD. More research about the role of OT is also required with regard to the prevention of the non-genetic intergenerational transmission of BPD. Clarifying the role of OT in BPD may also benefit from research in non-human animals targeting the interaction between early adversity and OT availability more directly. PRACTITIONER POINTS The study of oxytocin can contribute to the understanding of the neurobiology of borderline personality disorder. Oxytocin is critically involved in attachment security, and methylation of the oxytocin receptor may play a role in the epigenetic modulation of early adversity. The intranasal application of oxytocin may be a useful therapeutic adjunct to psychotherapy. Insecure attachment and childhood adversity may produce differential neurobiological effects on the oxytocinergic system in borderline personality disorder. There is insufficient knowledge of how oxytocin interacts with vasopressin, testosterone, dopamine, and serotonin, which are also important key players in the experience of social reward and stress responsivity. It is unclear whether or not oxytocin could be beneficial in preventing the intergenerational (non-genetic) transmission of borderline personality traits.
Collapse
Affiliation(s)
- Martin Brüne
- Division of Cognitive Neuropsychiatry and Psychiatric Preventive Medicine, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital, Ruhr-University Bochum, Germany
| |
Collapse
|
47
|
Somato-axodendritic release of oxytocin into the brain due to calcium amplification is essential for social memory. J Physiol Sci 2015; 66:275-82. [PMID: 26586001 PMCID: PMC4893072 DOI: 10.1007/s12576-015-0425-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 10/28/2015] [Indexed: 01/12/2023]
Abstract
Oxytocin (OT) is released into the brain from the cell soma, axons, and dendrites of neurosecretory cells in the hypothalamus. Locally released OT can activate OT receptors, form inositol-1,4,5-trisphosphate and elevate intracellular free calcium (Ca2+) concentrations [(Ca2+)i] in self and neighboring neurons in the hypothalamus, resulting in further OT release: i.e., autocrine or paracrine systems of OT-induced OT release. CD38-dependent cyclic ADP-ribose (cADPR) is also involved in this autoregulation by elevating [Ca2+]i via Ca2+ mobilization through ryanodine receptors on intracellular Ca2+ pools that are sensitive to both Ca2+ and cADPR. In addition, it has recently been reported that heat stimulation and hyperthermia enhance [Ca2+]i increases by Ca2+ influx, probably through TRPM2 cation channels, suggesting that cADPR and TRPM2 molecules act as Ca2+ signal amplifiers. Thus, OT release is not simply due to depolarization–secretion coupling. Both of these molecules play critical roles not only during labor and milk ejection in reproductive females, but also during social behavior in daily life in both genders. This was clearly demonstrated in CD38 knockout mice in that social behavior was impaired by reduction of [Ca2+]i elevation and subsequent OT secretion. Evidence for the associations of CD38 with social behavior and psychiatric disorder is discussed, especially in subjects with autism spectrum disorder.
Collapse
|
48
|
Oxytocin selectively modulates brain response to stimuli probing social synchrony. Neuroimage 2015; 124:923-930. [PMID: 26455794 DOI: 10.1016/j.neuroimage.2015.09.066] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 09/30/2015] [Accepted: 09/30/2015] [Indexed: 12/28/2022] Open
Abstract
The capacity to act collectively within groups has led to the survival and thriving of Homo sapiens. A central group collaboration mechanism is "social synchrony," the coordination of behavior during joint action among affiliative members, which intensifies under threat. Here, we tested brain response to vignettes depicting social synchrony among combat veterans trained for coordinated action and following life-threatening group experience, versus controls, as modulated by oxytocin (OT), a neuropeptide supporting social synchrony. Using a randomized, double-blind, within-subject design, 40 combat-trained and control male veterans underwent magnetoencephalography (MEG) twice following OT/placebo administration while viewing two social vignettes rated as highly synchronous: pleasant male social gathering and coordinated unit during combat. Both vignettes activated a wide response across the social brain in the alpha band; the combat scene triggered stronger activations. Importantly, OT effects were modulated by prior experience. Among combat veterans, OT attenuated the increased response to combat stimuli in the posterior superior temporal sulcus (pSTS) - a hub of social perception, action observation, and mentalizing - and enhanced activation in the inferior parietal lobule (IPL) to the pleasant social scene. Among controls, OT enhanced inferior frontal gyrus (IFG) response to combat cues, demonstrating selective OT effects on mirror-neuron and mentalizing networks. OT-enhanced mirror network activity was dampened in veterans reporting higher posttraumatic symptoms. Results demonstrate that the social brain responds online, via modulation of alpha rhythms, to stimuli probing social synchrony, particularly those involving threat to survival, and OT's enhancing versus anxiolytic effects are sensitive to salient experiences within social groups.
Collapse
|
49
|
Yamasue H. Using endophenotypes to examine molecules related to candidate genes as novel therapeutics: The “endophenotype-associated surrogate endpoint (EASE)” concept. Neurosci Res 2015; 99:1-7. [DOI: 10.1016/j.neures.2015.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 05/25/2015] [Accepted: 05/25/2015] [Indexed: 01/16/2023]
|
50
|
Watanabe T, Kuroda M, Kuwabara H, Aoki Y, Iwashiro N, Tatsunobu N, Takao H, Nippashi Y, Kawakubo Y, Kunimatsu A, Kasai K, Yamasue H. Clinical and neural effects of six-week administration of oxytocin on core symptoms of autism. Brain 2015; 138:3400-12. [PMID: 26336909 DOI: 10.1093/brain/awv249] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 07/13/2015] [Indexed: 12/21/2022] Open
Abstract
Autism spectrum disorder is a prevalent neurodevelopmental disorder with no established pharmacological treatment for its core symptoms. Although previous literature has shown that single-dose administration of oxytocin temporally mitigates autistic social behaviours in experimental settings, it remains in dispute whether such potentially beneficial responses in laboratories can result in clinically positive effects in daily life situations, which are measurable only in long-term observations of individuals with the developmental disorder undergoing continual oxytocin administration. Here, to address this issue, we performed an exploratory, randomized, double-blind, placebo-controlled, crossover trial including 20 high-functional adult males with autism spectrum disorder. Data obtained from 18 participants who completed the trial showed that 6-week intranasal administration of oxytocin significantly reduced autism core symptoms specific to social reciprocity, which was clinically evaluated by Autism Diagnostic Observation Scale (P = 0.034, PFDR < 0.05, Cohen's d = 0.78). Critically, the improvement of this clinical score was accompanied by oxytocin-induced enhancement of task-independent resting-state functional connectivity between anterior cingulate cortex and dorso-medial prefrontal cortex (rho = -0.60, P = 0.011), which was measured by functional magnetic resonance imaging. Moreover, using the same social-judgement task as used in our previous single-dose oxytocin trial, we confirmed that the current continual administration also significantly mitigated behavioural and neural responses during the task, both of which were originally impaired in autistic individuals (judgement tendency: P = 0.019, d = 0.62; eye-gaze effect: P = 0.03, d = 0.56; anterior cingulate activity: P = 0.00069, d = 0.97; dorso-medial prefrontal activity: P = 0.0014, d = 0.92; all, PFDR < 0.05). Furthermore, despite its longer administration, these effect sizes of the 6-week intervention were not larger than those seen in our previous single-dose intervention. These findings not only provide the evidence for clinically beneficial effects of continual oxytocin administration on the core social symptoms of autism spectrum disorder with suggesting its underlying biological mechanisms, but also highlight the necessity to seek optimal regimens of continual oxytocin treatment in future studies.
Collapse
Affiliation(s)
- Takamitsu Watanabe
- 1 Department of Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan 2 Department of Physiology, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan 3 Institute of Cognitive Neuroscience, University College London, 17 Queen Square, London, WC1N 3AR, UK
| | - Miho Kuroda
- 4 Department of Child Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Hitoshi Kuwabara
- 4 Department of Child Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yuta Aoki
- 1 Department of Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Norichika Iwashiro
- 1 Department of Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Natsubori Tatsunobu
- 1 Department of Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Hidemasa Takao
- 5 Department of Radiology, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yasumasa Nippashi
- 5 Department of Radiology, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yuki Kawakubo
- 4 Department of Child Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Akira Kunimatsu
- 5 Department of Radiology, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kiyoto Kasai
- 1 Department of Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Hidenori Yamasue
- 1 Department of Neuropsychiatry, School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan 6 Japan Science and Technology Agency, CREST, 5 Sambancho, Chiyoda-ku, Tokyo 102-0075, Japan
| |
Collapse
|