1
|
Nerio-Morales LK, Boender AJ, Young LJ, Lamprea MR, Smith AS. Limbic oxytocin receptor expression alters molecular signaling and social avoidance behavior in female prairie voles ( Microtus ochrogaster). Front Neurosci 2024; 18:1409316. [PMID: 39081850 PMCID: PMC11286410 DOI: 10.3389/fnins.2024.1409316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Introduction The social defeat paradigm is the most representative animal model to study social anxiety disorder (SAD) and its underlying neuronal mechanisms. We have previously reported that defeat progressively reduces oxytocin receptors (OXTR) in limbic regions of the brain over an eight-week period in female prairie voles (Microtus ochrogaster). Oxytocin receptors activate the mitogen-activated protein kinase (MAPK) pathway, which has been previously associated with the anxiolytic effects of oxytocin. Here, we assessed the functional significance of OXTR in stress-induced social avoidance and the response of the MAPK signaling pathway in the nucleus accumbens (NAc), anterior cingulate cortex (ACC), and basolateral amygdala (BLA) of female prairie voles. Methods In experiment 1, Sexually naïve adult female prairie voles were defeated for three consecutive days and tested a week after for social preference/avoidance (SPA) test. Control subjects were similarly handled without defeat conditioning. In experiment 2, sexually and stress naïve adult female prairie voles were bilaterally injected into the NAc, ACC, or the BLA with a CRISPR/Cas9 virus targeting the Oxtr coding sequence to induce OXTR knockdown. Two weeks post-surgery, subjects were tested for SPA behavior. Viral control groups were similarly handled but injected with a control virus. A subgroup of animals from each condition in both experiments were similarly treated and euthanized without being tested for SPA behavior. Brains were harvested for OXTR autoradiography, western blot analysis of MAPK proteins and quantification of local oxytocin content in the NAc, BLA, ACC, and PVN through ELISA. Results Social defeat reduced OXTR binding in the NAc and affected MAPK pathway activity and oxytocin availability. These results were region-specific and sensitive to exposure to the SPA test. Additionally, OXTR knockdown in the NAc, ACC, and BLA induced social avoidance and decreased basal MAPK activity in the NAc. Finally, we found that OXTR knockdown in these regions was associated with less availability of oxytocin in the PVN. Conclusion Dysregulation of the oxytocin system and MAPK signaling pathway in the NAc, ACC, and BLA are important in social behavior disruptions in female voles. This dysregulation could, therefore, play an important role in the etiology of SAD in women.
Collapse
Affiliation(s)
- Lina K. Nerio-Morales
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| | - Arjen J. Boender
- Department of Psychiatry and Behavioral Sciences, Center for Translational Social Neuroscience, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Larry J. Young
- Department of Psychiatry and Behavioral Sciences, Center for Translational Social Neuroscience, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Marisol R. Lamprea
- Department of Psychology, School of Human Sciences, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Adam S. Smith
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, United States
- Program in Neuroscience, School of Pharmacy, University of Kansas, Lawrence, KS, United States
| |
Collapse
|
2
|
Zhou H, Zhu R, Xia Y, Zhang X, Wang Z, Lorimer GH, Ghiladi RA, Bayram H, Wang J. Neuropeptides affecting social behavior in mammals: Oxytocin. Peptides 2024; 177:171223. [PMID: 38626843 DOI: 10.1016/j.peptides.2024.171223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/24/2024] [Accepted: 04/13/2024] [Indexed: 04/30/2024]
Abstract
Oxytocin (OXT), a neuropeptide consisting of only nine amino acids, is synthesized in the paraventricular and supraoptic nuclei of the hypothalamus. Although OXT is best known for its role in lactation and parturition, recent research has shown that it also has a significant impact on social behaviors in mammals. However, a comprehensive review of this topic is still lacking. In this paper, we systematically reviewed the effects of OXT on social behavior in mammals. These effects of OXT from the perspective of five key behavioral dimensions were summarized: parental behavior, anxiety, aggression, attachment, and empathy. To date, researchers have agreed that OXT plays a positive regulatory role in a wide range of social behaviors, but there have been controversially reported results. In this review, we have provided a detailed panorama of the role of OXT in social behavior and, for the first time, delved into the underlying regulatory mechanisms, which may help better understand the multifaceted role of OXT. Levels of OXT in previous human studies were also summarized to provide insights for diagnosis of mental disorders.
Collapse
Affiliation(s)
- Hong Zhou
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan, Hubei 430068, China; International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei 430068, China; National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, Hubei 430068, China
| | - Rui Zhu
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan, Hubei 430068, China; International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei 430068, China; National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, Hubei 430068, China
| | - Yuqing Xia
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan, Hubei 430068, China; International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei 430068, China; National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, Hubei 430068, China
| | - Xinming Zhang
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan, Hubei 430068, China; International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei 430068, China; National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, Hubei 430068, China
| | - Zixu Wang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, Hubei 430068, China
| | | | - Reza A Ghiladi
- Department of Chemistry, North Carolina State University, Raleigh, NC 27695, USA
| | - Hasan Bayram
- Department of Pulmonary Medicine, Koç University School of Medicine, Istanbul 34450, Turkey
| | - Jun Wang
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei University of Technology, Wuhan, Hubei 430068, China; International Center for Redox Biology & Precision Medicine of Hubei Province, Hubei University of Technology, Wuhan, Hubei 430068, China; National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, Hubei 430068, China.
| |
Collapse
|
3
|
Winter J, Meyer M, Berger I, Royer M, Bianchi M, Kuffner K, Peters S, Stang S, Langgartner D, Hartmann F, Schmidtner AK, Reber SO, Bosch OJ, Bludau A, Slattery DA, van den Burg EH, Jurek B, Neumann ID. Chronic oxytocin-driven alternative splicing of Crfr2α induces anxiety. Mol Psychiatry 2023; 28:4742-4755. [PMID: 34035479 PMCID: PMC10914602 DOI: 10.1038/s41380-021-01141-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/25/2021] [Accepted: 04/20/2021] [Indexed: 12/26/2022]
Abstract
The neuropeptide oxytocin (OXT) has generated considerable interest as potential treatment for psychiatric disorders, including anxiety and autism spectrum disorders. However, the behavioral and molecular consequences associated with chronic OXT treatment and chronic receptor (OXTR) activation have scarcely been studied, despite the potential therapeutic long-term use of intranasal OXT. Here, we reveal that chronic OXT treatment over two weeks increased anxiety-like behavior in rats, with higher sensitivity in females, contrasting the well-known anxiolytic effect of acute OXT. The increase in anxiety was transient and waned 5 days after the infusion has ended. The behavioral effects of chronic OXT were paralleled by activation of an intracellular signaling pathway, which ultimately led to alternative splicing of hypothalamic corticotropin-releasing factor receptor 2α (Crfr2α), an important modulator of anxiety. In detail, chronic OXT shifted the splicing ratio from the anxiolytic membrane-bound (mCRFR2α) form of CRFR2α towards the soluble CRFR2α (sCRFR2α) form. Experimental induction of alternative splicing mimicked the anxiogenic effects of chronic OXT, while sCRFR2α-knock down reduced anxiety-related behavior of male rats. Furthermore, chronic OXT treatment triggered the release of sCRFR2α into the cerebrospinal fluid with sCRFR2α levels positively correlating with anxiety-like behavior. In summary, we revealed that the shifted splicing ratio towards expression of the anxiogenic sCRFR2α underlies the adverse effects of chronic OXT treatment on anxiety.
Collapse
Affiliation(s)
- Julia Winter
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Magdalena Meyer
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Ilona Berger
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Melanie Royer
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Marta Bianchi
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Kerstin Kuffner
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Sebastian Peters
- Department of Neurology, University Hospital Regensburg, Regensburg, Germany
| | - Simone Stang
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Dominik Langgartner
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, University of Ulm, Ulm, Germany
| | - Finn Hartmann
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Anna K Schmidtner
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Stefan O Reber
- Laboratory for Molecular Psychosomatics, Department of Psychosomatic Medicine and Psychotherapy, University of Ulm, Ulm, Germany
| | - Oliver J Bosch
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Anna Bludau
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - David A Slattery
- Laboratory of Translational Psychiatry, Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University of Frankfurt, Frankfurt am Main, Germany
| | - Erwin H van den Burg
- Center for Psychiatric Neurosciences, University Hospital Lausanne, Lausanne, Switzerland
| | - Benjamin Jurek
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany
| | - Inga D Neumann
- Department of Behavioural and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
4
|
Neumann ID. Monitoring oxytocin signaling in the brain: More than a love story. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2023; 16:100206. [PMID: 38108033 PMCID: PMC10724740 DOI: 10.1016/j.cpnec.2023.100206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 12/19/2023] Open
Abstract
More than any other neuropeptide, oxytocin (OXT) is attracting the attention of neurobiologists, psychologists, psychiatrists, evolutionary biologists and even economists. It is often called a "love hormone" due to its many prosocial functions described in vertebrates including mammals and humans, especially its ability to support "bonding behaviour". Oxytocin plays an important role in female reproduction, as it promotes labour during parturition, enables milk ejection in lactation and is essential for related reproductive behaviours. Therefore, it particularly attracts the interest of many female researchers. In this short narrative review I was invited to provide a personal overview on my scientific journey closely linked to my research on the brain OXT system and the adventures associated with starting my research career behind the Iron Curtain.
Collapse
Affiliation(s)
- Inga D. Neumann
- Department of Behavioural and Molecular Neurobiology, Regensburg Centre of Neurosciences, University of Regensburg, Regensburg, Germany
| |
Collapse
|
5
|
Pandamooz S, Salehi MS, Jurek B, Meinung CP, Azarpira N, Dianatpour M, Neumann ID. Oxytocin Receptor Expression in Hair Follicle Stem Cells: A Promising Model for Biological and Therapeutic Discovery in Neuropsychiatric Disorders. Stem Cell Rev Rep 2023; 19:2510-2524. [PMID: 37548806 DOI: 10.1007/s12015-023-10603-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2023] [Indexed: 08/08/2023]
Abstract
The intricate nature of the human brain and the limitations of existing model systems to study molecular and cellular causes of neuropsychiatric disorders represent a major challenge for basic research. The promising progress in patient-derived stem cell technology and in our knowledge on the role of the brain oxytocin (OXT) system in health and disease offer new possibilities in that direction. In this study, the rat hair follicle stem cells (HFSCs) were isolated and expanded in vitro. The expression of oxytocin receptors (OXTR) was evaluated in these cells. The cellular viability was assessed 12 h post stimulation with OXT. The activation of OXTR-coupled intracellular signaling cascades, following OXT treatment was determined. Also, the influence of OXT on neurite outgrowth and cytoskeletal rearrangement were defined. The assessment of OXTR protein expression revealed this receptor is expressed abundantly in HFSCs. As evidenced by the cell viability assay, no adverse or cytotoxic effects were detected following 12 h treatment with different concentrations of OXT. Moreover, OXTR stimulation by OXT resulted in ERK1/2, CREB, and eEF2 activation, neurite length alterations, and cytoskeletal rearrangements that reveal the functionality of this receptor in HFSCs. Here, we introduced the rat HFSCs as an easy-to-obtain stem cell model that express functional OXTR. This cell-based model can contribute to our understanding of the progression and treatment of neuropsychiatric disorders with oxytocinergic system deficiency.
Collapse
Affiliation(s)
- Sareh Pandamooz
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Molecular and Behavioural Neurobiology, University of Regensburg, Regensburg, Germany
| | - Mohammad Saied Salehi
- Department of Molecular and Behavioural Neurobiology, University of Regensburg, Regensburg, Germany.
- Clinical Neurology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Benjamin Jurek
- Department of Molecular and Behavioural Neurobiology, University of Regensburg, Regensburg, Germany
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Carl-Philipp Meinung
- Department of Molecular and Behavioural Neurobiology, University of Regensburg, Regensburg, Germany
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Dianatpour
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Inga D Neumann
- Department of Molecular and Behavioural Neurobiology, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
6
|
Jurek B, Denk L, Schäfer N, Salehi MS, Pandamooz S, Haerteis S. Oxytocin accelerates tight junction formation and impairs cellular migration in 3D spheroids: evidence from Gapmer-induced exon skipping. Front Cell Neurosci 2022; 16:1000538. [PMID: 36263085 PMCID: PMC9574052 DOI: 10.3389/fncel.2022.1000538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Oxytocin (OXT) is a neuropeptide that has been associated with neurological diseases like autism, a strong regulating activity on anxiety and stress-related behavior, physiological effects during pregnancy and parenting, and various cellular effects in neoplastic tissue. In this study, we aimed to unravel the underlying mechanism that OXT employs to regulate cell-cell contacts, spheroid formation, and cellular migration in a 3D culture model of human MLS-402 cells. We have generated a labeled OXT receptor (OXTR) overexpressing cell line cultivated in spheroids that were treated with the OXTR agonists OXT, Atosiban, and Thr4-Gly7-oxytocin (TGOT); with or without a pre-treatment of antisense oligos (Gapmers) that induce exon skipping in the human OXTR gene. This exon skipping leads to the exclusion of exon 4 and therefore a receptor that lost its intracellular G-protein-binding domain. Sensitive digital PCR (dPCR) provided us with the means to differentiate between wild type and truncated OXTR in our cellular model. OXTR truncation differentially activated intracellular signaling cascades related to cell-cell attachment and proliferation like Akt, ERK1/2-RSK1/2, HSP27, STAT1/5, and CREB, as assessed by a Kinase Profiler Assay. Digital and transmission electron microscopy revealed increased tight junction formation and well-organized cellular protrusions into an enlarged extracellular space after OXT treatment, resulting in increased cellular survival. In summary, OXT decreases cellular migration but increases cell-cell contacts and therefore improves nutrient supply. These data reveal a novel cellular effect of OXT that might have implications for degenerating CNS diseases and tumor formation in various tissues.
Collapse
Affiliation(s)
- Benjamin Jurek
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany
- Research Group Neurobiology of Stress Resilience, Max Planck Institute of Psychiatry, Munich, Germany
| | - Lucia Denk
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany
| | - Nicole Schäfer
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany
- Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB), Bio Park 1, University of Regensburg, Regensburg, Germany
| | - Mohammad Saied Salehi
- Clinical Neurology Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Sareh Pandamooz
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Silke Haerteis
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Regensburg, Germany
- *Correspondence: Silke Haerteis
| |
Collapse
|
7
|
Dahchour A. Anxiolytic and antidepressive potentials of rosmarinic acid: A review with a focus on antioxidant and anti-inflammatory effects. Pharmacol Res 2022; 184:106421. [PMID: 36096427 DOI: 10.1016/j.phrs.2022.106421] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 10/14/2022]
Abstract
Depression and anxiety are the most prevalent neuropsychiatric disorders that have emerged as global health concerns. Anxiolytic and antidepressant drugs, such as benzodiazepines, selective serotonin reuptake inhibitors, monoamine oxidase inhibitors, and tricyclics, are the first line used in treating anxiety and depression. Although these drugs lack efficacy and have a delayed response time and numerous side effects, their widespread abuse and market continue to grow. Over time, traditional practices using natural and phytochemicals as alternative therapies to chemical drugs have emerged to treat many pathological conditions, including anxiety and depression. Recent preclinical studies have demonstrated that the phenolic compound, rosmarinic acid, is effective against several neuropsychiatric disorders, including anxiety and depression. In addition, rosmarinic acid showed various pharmacological effects, such as cardioprotective, hepatoprotective, lung protective, antioxidant, anti-inflammatory, and neuroprotective effects. However, the potentialities of the use of rosmarinic acid in the treatment of nervous system-related disorders, such as anxiety and depression, are less or not yet reviewed. Therefore, the purpose of this review was to present several preclinical and clinical studies, when available, from different databases investigating the effects of rosmarinic acid on anxiety and depression. These studies showed that rosmarinic acid produces advantageous effects on anxiety and depression through its powerful antioxidant and anti-inflammatory properties. This review will examine and discuss the possibility that the anxiolytic and anti-depressive effects of rosmarinic acid could be associated with its potent antioxidant and anti-inflammatory activities.
Collapse
Affiliation(s)
- Abdelkader Dahchour
- Clinical Neurosciences Laboratory, Faculty of Medicine and Pharmacy. Department of Biology, Faculty of Sciences, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco.
| |
Collapse
|
8
|
Bridging skin, brain, and behavior to understand pleasurable social touch. Curr Opin Neurobiol 2022; 73:102527. [PMID: 35453001 DOI: 10.1016/j.conb.2022.102527] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 02/09/2022] [Accepted: 02/15/2022] [Indexed: 02/07/2023]
Abstract
Social touch-the affiliative skin-to-skin contact between individuals-can rapidly evoke emotions of comfort, pleasure, or calm, and is essential for mental and physical well-being. Physical isolation from social support can be devastating. During the COVID-19 pandemic, we observed a global increase in suicidal ideation, anxiety, domestic violence, and worsening of pre-existing physical conditions, alerting society to our need to understand the neurobiology of social touch and how it promotes normal health. Gaining a mechanistic understanding of how sensory neuron stimulation induces pleasure, calm, and analgesia may reveal untapped therapeutic targets in the periphery for treatment of anxiety and depression, as well as social disorders and traumas in which social touch becomes aversive. Bridging the gap between stimulation in the skin and positive affect in the brain-especially during naturally occurring social touch behaviors-remains a challenge to the field. However, with advances in mouse genetics, behavioral quantification, and brain imaging approaches to measure neuronal firing and neurochemical release, completing this mechanistic picture may be on the horizon. Here, we summarize some exciting new findings about social touch in mammals, emphasizing both the peripheral and central nervous systems, with attempts to bridge the gap between external stimulation and internal representations in the brain.
Collapse
|
9
|
Wang F, Yin XS, Lu J, Cen C, Wang Y. Phosphorylation-dependent positive feedback on the oxytocin receptor through the kinase PKD1 contributes to long-term social memory. Sci Signal 2022; 15:eabd0033. [PMID: 35104164 DOI: 10.1126/scisignal.abd0033] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Social memory enables one to recognize and distinguish specific individuals. It is fundamental to social behaviors that can be mediated by the oxytocin receptor (OXTR), such as forming relationships. We investigated the molecular regulation and function of OXTR in animal behavior involving social memory. We found that Ser261 in OXTR was phosphorylated by protein kinase D1 (PKD1). Neuronal Ca2+ signaling and behavior analyses revealed that rats expressing a mutated form of OXTR that cannot be phosphorylated at this residue (OXTR S261A) in the medial amygdala (MeA) exhibited impaired long-term social memory (LTSM). Blocking the phosphorylation of wild-type OXTR in the MeA using an interfering peptide in rats or through conditional knockout of Pkd1 in mice reduced social memory retention, whereas expression of a phosphomimetic mutant of OXTR rescued it. In HEK293A cells, the PKD1-mediated phosphorylation of OXTR promoted its binding to Gq protein and, in turn, OXTR-mediated phosphorylation of PKD1, indicating a positive feedback loop. In addition, OXTR with a single-nucleotide polymorphism found in humans (rs200362197), which has a mutation in the conserved recognition region in the PKD1 phosphorylation site, showed impaired activation and signaling in vitro and in HEK293A cells similar to that of the S216A mutant. Our findings describe a phosphoregulatory loop for OXTR and its critical role in social behavior that might be further explored in associated disorders.
Collapse
Affiliation(s)
- Fei Wang
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute; Key Lab for Neuroscience, Ministry of Education of China and National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China.,Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xiang-Sha Yin
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute; Key Lab for Neuroscience, Ministry of Education of China and National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China
| | - Jie Lu
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute; Key Lab for Neuroscience, Ministry of Education of China and National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China
| | - Cheng Cen
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute; Key Lab for Neuroscience, Ministry of Education of China and National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China
| | - Yun Wang
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute; Key Lab for Neuroscience, Ministry of Education of China and National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100083, China.,PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| |
Collapse
|
10
|
Ghazy AA, Soliman OA, Elbahnasi AI, Alawy AY, Mansour AM, Gowayed MA. Role of Oxytocin in Different Neuropsychiatric, Neurodegenerative, and Neurodevelopmental Disorders. Rev Physiol Biochem Pharmacol 2022; 186:95-134. [PMID: 36416982 DOI: 10.1007/112_2022_72] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Oxytocin has recently gained significant attention because of its role in the pathophysiology and management of dominant neuropsychiatric disorders. Oxytocin, a peptide hormone synthesized in the hypothalamus, is released into different brain regions, acting as a neurotransmitter. Receptors for oxytocin are present in many areas of the brain, including the hypothalamus, amygdala, and nucleus accumbens, which have been involved in the pathophysiology of depression, anxiety, schizophrenia, autism, Alzheimer's disease, Parkinson's disease, and attention deficit hyperactivity disorder. Animal studies have spotlighted the role of oxytocin in social, behavioral, pair bonding, and mother-infant bonding. Furthermore, oxytocin protects fetal neurons against injury during childbirth and affects various behaviors, assuming its possible neuroprotective characteristics. In this review, we discuss some of the concepts and mechanisms related to the role of oxytocin in the pathophysiology and management of some neuropsychiatric, neurodegenerative, and neurodevelopmental disorders.
Collapse
Affiliation(s)
- Aya A Ghazy
- Department of Clinical Pharmacy, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Omar A Soliman
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Aya I Elbahnasi
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Aya Y Alawy
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Amira Ma Mansour
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Mennatallah A Gowayed
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt.
| |
Collapse
|
11
|
Structure-function relationships of the disease-linked A218T oxytocin receptor variant. Mol Psychiatry 2022; 27:907-917. [PMID: 34980886 PMCID: PMC9054668 DOI: 10.1038/s41380-021-01241-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/24/2021] [Accepted: 07/15/2021] [Indexed: 12/20/2022]
Abstract
Various single nucleotide polymorphisms (SNPs) in the oxytocin receptor (OXTR) gene have been associated with behavioral traits, autism spectrum disorder (ASD) and other diseases. The non-synonymous SNP rs4686302 results in the OXTR variant A218T and has been linked to core characteristics of ASD, trait empathy and preterm birth. However, the molecular and intracellular mechanisms underlying those associations are still elusive. Here, we uncovered the molecular and intracellular consequences of this mutation that may affect the psychological or behavioral outcome of oxytocin (OXT)-treatment regimens in clinical studies, and provide a mechanistic explanation for an altered receptor function. We created two monoclonal HEK293 cell lines, stably expressing either the wild-type or A218T OXTR. We detected an increased OXTR protein stability, accompanied by a shift in Ca2+ dynamics and reduced MAPK pathway activation in the A218T cells. Combined whole-genome and RNA sequencing analyses in OXT-treated cells revealed 7823 differentially regulated genes in A218T compared to wild-type cells, including 429 genes being associated with ASD. Furthermore, computational modeling provided a molecular basis for the observed change in OXTR stability suggesting that the OXTR mutation affects downstream events by altering receptor activation and signaling, in agreement with our in vitro results. In summary, our study provides the cellular mechanism that links the OXTR rs4686302 SNP with genetic dysregulations associated with aspects of ASD.
Collapse
|
12
|
Stanić D, Oved K, Israel-Elgali I, Jukić M, Batinić B, Puškaš N, Shomron N, Gurwitz D, Pešić V. Synergy of oxytocin and citalopram in modulating Itgb3/Chl1 interplay: Relevance to sensitivity to SSRI therapy. Psychoneuroendocrinology 2021; 129:105234. [PMID: 33930757 DOI: 10.1016/j.psyneuen.2021.105234] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 11/16/2022]
Abstract
Intranasal treatment with oxytocin showed beneficial effects in post-traumatic stress disorder and autism spectrum disorders; however, it was not investigated as much in depression. Keeping in mind the favorable effects of oxytocin on animal models of anxiety and depression, we postulated that synergy between prescribed first choice drugs, selective serotonin reuptake inhibitors (SSRIs) and oxytocin could improve the treatment outcome compared with SSRI monotherapy. Our previous in vitro genome-wide transcriptomic study on human lymphoblastoid cell lines exposed to paroxetine resulted in increase of integrin β3 (ITGB3) gene expression, and further, ITGB3/CHL1 expression ratio was hypothesized to influence the sensitivity to SSRIs. The aim of this report was to explore molecular mechanisms behind the antidepressant-like oxytocin effect, alone and in synergy with citalopram, on behavioral and molecular level in corticosterone treated rats, a paradigm used to model anxiety and depression in animals. Oxytocin treatment (1) ameliorated corticosterone-induced reduction of neurogenesis and number of parvalbumin-positive interneurons in the hippocampal CA1 region, (2) enhanced anxiolytic- and antidepressant-like effects of citalopram in the open field test, and (3) the SSRI/oxytocin synergy persisted in reversing the reduction of the Itgb3 gene expression and increased Itgb3/Chl1 ratio in the prefrontal cortices. These results support the existence of synergy between citalopram and oxytocin in reversing the molecular and behavioral changes induced by corticosterone treatment and point to possible molecular mechanisms behind antidepressant-like effect of oxytocin.
Collapse
Affiliation(s)
- Dušanka Stanić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11121 Belgrade, Serbia
| | - Keren Oved
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ifat Israel-Elgali
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| | - Marin Jukić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11121 Belgrade, Serbia; Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Bojan Batinić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11121 Belgrade, Serbia
| | - Nela Puškaš
- Department of Histology and Embryology, Faculty of Medicine, University of Belgrade, Serbia
| | - Noam Shomron
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| | - David Gurwitz
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| | - Vesna Pešić
- Department of Physiology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11121 Belgrade, Serbia.
| |
Collapse
|
13
|
Cell proliferation and anti-oxidant effects of oxytocin and oxytocin receptors: role of extracellular signal-regulating kinase in astrocyte-like cells. Endocr Regul 2021; 54:172-182. [PMID: 32857718 DOI: 10.2478/enr-2020-0020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Oxytocin (OXT) participates in various physiological functions ranging from reproduction to social and non-social behaviors. Recent studies indicate that OXT affects cell growth and metabolism. Here we characterized the growth stimulating and antioxidant actions of OXT and of OXT receptors (OXTR) in a glial cell-line (U-87MG). METHODS We developed an OXTR-knockdown cell-line (U-87MG KD) to establish the receptor specificity of OXT's actions, and the impact of lacking OXTR on growth and survival in glial cells. The role Extracellular-Signal Regulated Kinases (ERK1/2) on glial cell protection against consequences of oxidative stress, and cell proliferation was investigated. RESULTS In U-87MG cells, OXT stimulated cell proliferation and increased ERK1/2 phosphorylation. The specific ERK1/2 inhibitor, PD098059, produced marked inhibition of cell proliferation, and antagonized the stimulating effect of OXT on ERK1/2 phosphorylation and on cell proliferation. Slower growth rates and lower levels of phosphorylated ERK1/2 were observed in OXTR-knockdown cells and in U-87MG cells treated with an OXTR antagonist (L-371,257). In addition to increasing cell proliferation, OXT significantly blunted the rise in reactive oxygen species induced by H2O2, and antagonized the reductions in cell viability induced by H2O2 and camptothecin. The cell protective and antioxidant actions of OXT in U-87MG cells were not observed in the OXTR-knockdown cells. CONCLUSION OXT stimulates the growth of astrocyte-like cells acting on OXTR via ERK1/2 phosphorylation. The protection against apoptosis and the antioxidant capacity of OXT may contribute to the observed increase in cell proliferation. Oxytocin and OXTR appear to be fundamental for cell growth and viability of glial cells.
Collapse
|
14
|
Che X, Cai J, Liu Y, Xu T, Yang J, Wu C. Oxytocin signaling in the treatment of drug addiction: Therapeutic opportunities and challenges. Pharmacol Ther 2021; 223:107820. [PMID: 33600854 DOI: 10.1016/j.pharmthera.2021.107820] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2021] [Indexed: 12/13/2022]
Abstract
Drug addiction is one of the leading causes of mortality worldwide. Despite great advances were achieved in understanding the neurobiology of drug addiction, the therapeutic options are severely limited, with poor effectiveness and serious side effects. The neuropeptide oxytocin (OXT) is well known for its effects on uterine contraction, sexual/maternal behaviors, social affiliation, stress and learning/memory by interacting with the OXT receptor and other neuromodulators. Emerging evidence suggests that the acute or chronic exposure to drugs can affect the OXT system. Additionally, OXT administration can ameliorate a wide range of abused drug-induced neurobehavioral changes. Overall, OXT not only suppresses drug reward in the binge stage of drug addiction, but also reduces stress responses and social impairments during the withdrawal stage and, finally, prevents drug/cue/stress-induced reinstatement. More importantly, clinical studies have also shown that OXT can exert beneficial effects on reducing substance use disorders of a series of drugs, such as heroin, cocaine, alcohol, cannabis and nicotine. Thus, the present review focuses on the role of OXT in treating drug addiction, including the preclinical and clinical therapeutic potential of OXT and its analogs on the neurobiological perspectives of drugs, to provide a better insight of the efficacy of OXT as a clinical addiction therapeutic agent.
Collapse
Affiliation(s)
- Xiaohang Che
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Screening of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Pharmacodynamics Evaluation of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Jialing Cai
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Yueyang Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Screening of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Pharmacodynamics Evaluation of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Tianyu Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Jingyu Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Screening of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Pharmacodynamics Evaluation of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China.
| | - Chunfu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Screening of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China; Key Laboratory of New Drug Pharmacodynamics Evaluation of Liaoning Province, Shenyang Pharmaceutical University, Shenyang, PR China.
| |
Collapse
|
15
|
Xu K, Wang M, Zhou W, Pu J, Wang H, Xie P. Chronic D-ribose and D-mannose overload induce depressive/anxiety-like behavior and spatial memory impairment in mice. Transl Psychiatry 2021; 11:90. [PMID: 33531473 PMCID: PMC7854712 DOI: 10.1038/s41398-020-01126-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/10/2020] [Accepted: 11/27/2020] [Indexed: 02/07/2023] Open
Abstract
The effects of different forms of monosaccharides on the brain remain unclear, though neuropsychiatric disorders undergo changes in glucose metabolism. This study assessed cell viability responses to five commonly consumed monosaccharides-D-ribose (RIB), D-glucose, D-mannose (MAN), D-xylose and L-arabinose-in cultured neuro-2a cells. Markedly decreased cell viability was observed in cells treated with RIB and MAN. We then showed that high-dose administration of RIB induced depressive- and anxiety-like behavior as well as spatial memory impairment in mice, while high-dose administration of MAN induced anxiety-like behavior and spatial memory impairment only. Moreover, significant pathological changes were observed in the hippocampus of high-dose RIB-treated mice by hematoxylin-eosin staining. Association analysis of the metabolome and transcriptome suggested that the anxiety-like behavior and spatial memory impairment induced by RIB and MAN may be attributed to the changes in four metabolites and 81 genes in the hippocampus, which is involved in amino acid metabolism and serotonin transport. In addition, combined with previous genome-wide association studies on depression, a correlation was found between the levels of Tnni3k and Tbx1 in the hippocampus and RIB induced depressive-like behavior. Finally, metabolite-gene network, qRT-PCR and western blot analysis showed that the insulin-POMC-MEK-TCF7L2 and MAPK-CREB-GRIN2A-CaMKII signaling pathways were respectively associated with RIB and MAN induced depressive/anxiety-like behavior and spatial memory impairment. Our findings clarified our understanding of the biological mechanisms underlying RIB and MAN induced depressive/anxiety-like behavior and spatial memory impairment in mice and highlighted the deleterious effects of high-dose RIB and MAN as long-term energy sources.
Collapse
Affiliation(s)
- Ke Xu
- grid.203458.80000 0000 8653 0555Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China ,grid.452206.7NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China ,grid.203458.80000 0000 8653 0555Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Mingyang Wang
- grid.452206.7NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China ,grid.203458.80000 0000 8653 0555Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China ,grid.203458.80000 0000 8653 0555College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Wei Zhou
- grid.452206.7NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China ,grid.203458.80000 0000 8653 0555Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Juncai Pu
- grid.452206.7NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China ,grid.203458.80000 0000 8653 0555Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Haiyang Wang
- grid.452206.7NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China ,grid.203458.80000 0000 8653 0555Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Peng Xie
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China. .,NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China. .,Institute of Neuroscience and Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China. .,Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
16
|
Sabihi S, Goodpaster C, Maurer S, Leuner B. GABA in the medial prefrontal cortex regulates anxiety-like behavior during the postpartum period. Behav Brain Res 2021; 398:112967. [PMID: 33075397 PMCID: PMC7722033 DOI: 10.1016/j.bbr.2020.112967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/02/2020] [Accepted: 10/07/2020] [Indexed: 01/22/2023]
Abstract
The postpartum period is commonly accompanied by emotional changes, which for many new mothers includes a reduction in anxiety. Previous research in rodents has shown that the postpartum attenuation in anxiety is dependent on offspring contact and has further implicated enhanced GABAergic neurotransmission as an underlying mechanism. However, the specific brain regions where GABA acts to regulate the offspring-induced reduction in postpartum anxiety requires further investigation. Here, we test the hypothesis that offspring interactions suppress anxiety-like behavior in postpartum female rats via GABA signaling in the medial prefrontal cortex (mPFC). Our results show a postpartum reduction in anxiety-like behavior, an effect which was abolished by localized infusion of the GABAA receptor antagonist bicuculline in the mPFC. We also show that activation of GABAA receptors in the mPFC by the agonist muscimol was effective in restoring anxiolyisis in mothers separated from their pups. Lastly, we show that heightened anxiety-like behavior in pup-separated mothers was accompanied by a lower number and percentage of activated GABAergic neurons within the mPFC. Together, these results suggest that mother-offspring interactions reduce anxiety-like behavior in postpartum females via GABAA neurotransmission in the mPFC and in doing so provide insight into mechanisms that may become dysfunctional in mothers who experience high postpartum anxiety.
Collapse
Affiliation(s)
- Sara Sabihi
- Department of Psychology, The Ohio State University Columbus, OH 43210, USA
| | - Caitlin Goodpaster
- Department of Psychology, The Ohio State University Columbus, OH 43210, USA
| | - Skyler Maurer
- Department of Psychology, The Ohio State University Columbus, OH 43210, USA
| | - Benedetta Leuner
- Department of Psychology, The Ohio State University Columbus, OH 43210, USA; Department of Neuroscience, The Ohio State University Columbus, OH 43210, USA.
| |
Collapse
|
17
|
Althammer F, Eliava M, Grinevich V. Central and peripheral release of oxytocin: Relevance of neuroendocrine and neurotransmitter actions for physiology and behavior. HANDBOOK OF CLINICAL NEUROLOGY 2021; 180:25-44. [PMID: 34225933 DOI: 10.1016/b978-0-12-820107-7.00003-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The hypothalamic neuropeptide oxytocin (OT) is critically involved in the modulation of socio-emotional behavior, sexual competence, and pain perception and anticipation. While intracellular signaling of OT and its receptor (OTR), as well as the functional connectivity of hypothalamic and extra-hypothalamic OT projections, have been recently explored, it remains elusive how one single molecule has pleotropic effects from cell proliferation all the way to modulation of complex cognitive processes. Moreover, there are astonishing species-dependent differences in the way OT regulates various sensory modalities such as touch, olfaction, and vision, which can be explained by differences in OTR expression in brain regions processing sensory information. Recent research highlights a small subpopulation of OT-synthesizing cells, namely, parvocellular cells, which merely constitute 1% of the total number of OT cells but act as "master cells' that regulate the activity of the entire OT system. In this chapter, we summarize the latest advances in the field of OT research with a particular focus on differences between rodents, monkeys and humans and highlight the main differences between OT and its "sister" peptide arginine-vasopressin, which often exerts opposite effects on physiology and behavior.
Collapse
Affiliation(s)
- Ferdinand Althammer
- Neuroscience Department, Center for Neuroinflammation and Cardiometabolic Diseases, Georgia State University, Atlanta, GA, United States
| | - Marina Eliava
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Valery Grinevich
- Department of Neuropeptide Research in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
18
|
Brain oxytocin: how puzzle stones from animal studies translate into psychiatry. Mol Psychiatry 2021; 26:265-279. [PMID: 32514104 PMCID: PMC7278240 DOI: 10.1038/s41380-020-0802-9] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/14/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023]
Abstract
The neuropeptide oxytocin has attracted great attention of the general public, basic neuroscience researchers, psychologists, and psychiatrists due to its profound pro-social, anxiolytic, and "anti-stress" behavioral and physiological effects, and its potential application for treatment of mental diseases associated with altered socio-emotional competence. During the last decade, substantial progress has been achieved in understanding the complex neurobiology of the oxytocin system, including oxytocinergic pathways, local release patterns, and oxytocin receptor distribution in the brain, as well as intraneuronal oxytocin receptor signaling. However, the picture of oxytocin actions remains far from being complete, and the central question remains: "How does a single neuropeptide exert such pleotropic actions?" Although this phenomenon, typical for many of about 100 identified neuropeptides, may emerge from the anatomical divergence of oxytocin neurons, their multiple central projections, distinct oxytocin-sensitive cell types in different brain regions, and multiple intraneuronal signaling pathways determining the specific cellular response, further basic studies are required. In conjunction, numerous reports on positive effects of intranasal application of oxytocin on human brain networks controlling socio-emotional behavior in health and disease require harmonic tandems of basic researchers and clinicians. During the COVID-19 crisis in 2020, oxytocin research seems central as question of social isolation-induced inactivation of the oxytocin system, and buffering effects of either activation of the endogenous system or intranasal application of synthetic oxytocin need to be thoroughly investigated.
Collapse
|
19
|
Paul A, Shakya A, Zaman MK. Assessment of acute and sub-chronic neurotoxicity of Morus alba L. fruits in rodents. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2020. [DOI: 10.1186/s43094-020-00110-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2023] Open
Abstract
Abstract
Background
Morus alba L. fruits are consumed since long for their nutritional and medicinal values. Although there were studies on the neuroprotective activity of the fruit extract, safety profile of the fruit extract is not yet explored as per the recommended standard guidelines over the central nervous system (CNS). The present work was aimed to assess the neurotoxicity profile of chemically characterized extract of M. alba L. fruits (MA) using validated OECD guidelines, i.e., 425 and 424 in rodents.
Results
Neurobehavioural parameters were examined for motor, sensory and behavioural responses using actophotometer, hot plate and light and dark box test, respectively as per OECD 424. Interestingly, no sign of mortality and/or adversity on mice treated per-orally with MA (2000 mg/kg) was observed during the limit test as per OECD 425. Further, rats treated with MA (1000, 300 and 100 mg/kg, p.o.) for 28 days, showed insignificant (p < 0.05) changes in body weight, food consumption, neurobehavioural responses, organ weights and biochemical, haematological and histopathological features when compared with vehicle-treated animals.
Conclusion
The outcome of findings suggests that MA is safe in acute oral as well as sub-chronic (28 days) administration in mice and rats respectively. MA (1000 mg/kg) did not pose any toxic sign and symptoms on neurobehavioural responses in rats even after 28 days repeated treatment in compliance with OECD 424.
Collapse
|
20
|
Wei J, Ma L, Ju P, Yang B, Wang YX, Chen J. Involvement of Oxytocin Receptor/Erk/MAPK Signaling in the mPFC in Early Life Stress-Induced Autistic-Like Behaviors. Front Cell Dev Biol 2020; 8:564485. [PMID: 33134294 PMCID: PMC7561716 DOI: 10.3389/fcell.2020.564485] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022] Open
Abstract
The neonatal or infant period is a critical stage for the development of brain neuroplasticity. Early life stresses in the neonatal period, including neonatal maternal separation (NMS), have adverse effects on an increased risk of psychiatric disorders in juveniles and adults. However, the underlying molecular mechanisms are not largely understood. Here, we found that juvenile rats subjected to 4 h daily NMS during postnatal days 1 to 20 exhibited autistic-like behavioral deficits without impairments in learning and memory functions. Molecular mechanism studies showed that oxytocin receptor (OXTR) in the medial prefrontal cortex of NMS rats was evidently downregulated when compared with control pups, especially in neurons. Erk/MAPK signaling, the downstream coupling signaling of OTXR, was also inhibited in NMS juvenile rats. Treatment with oxytocin could relieve NMS-induced social deficit behaviors and activated phosphorylation of Erk/MAPK signaling. Furthermore, medication with the inhibitor of H3K4 demethylase alleviated the abnormal behaviors in NMS rats and increased the expression of OXTR in the medial prefrontal cortex, which showed an epigenetic mechanism underlying social deficits induced by NMS. Taken together, these findings identified a molecular mechanism by which disruptions of mother-infant interactions influenced later displays of typical social behaviors and suggested the potential for NMS-driven epigenetic tuning of OXTR expression.
Collapse
Affiliation(s)
- Jinbao Wei
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,King's Lab, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China.,Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Institute of Wudang Traditional Chinese Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Le Ma
- King's Lab, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Peijun Ju
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Beibei Yang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yong-Xiang Wang
- King's Lab, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Jinghong Chen
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
21
|
Albert-Gascó H, Ros-Bernal F, Castillo-Gómez E, Olucha-Bordonau FE. MAP/ERK Signaling in Developing Cognitive and Emotional Function and Its Effect on Pathological and Neurodegenerative Processes. Int J Mol Sci 2020; 21:E4471. [PMID: 32586047 PMCID: PMC7352860 DOI: 10.3390/ijms21124471] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/14/2020] [Accepted: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
The signaling pathway of the microtubule-associated protein kinase or extracellular regulated kinase (MAPK/ERK) is a common mechanism of extracellular information transduction from extracellular stimuli to the intracellular space. The transduction of information leads to changes in the ongoing metabolic pathways and the modification of gene expression patterns. In the central nervous system, ERK is expressed ubiquitously, both temporally and spatially. As for the temporal ubiquity, this signaling system participates in three key moments: (i) Embryonic development; (ii) the early postnatal period; and iii) adulthood. During embryonic development, the system is partly responsible for the patterning of segmentation in the encephalic vesicle through the FGF8-ERK pathway. In addition, during this period, ERK directs neurogenesis migration and the final fate of neural progenitors. During the early postnatal period, ERK participates in the maturation process of dendritic trees and synaptogenesis. During adulthood, ERK participates in social and emotional behavior and memory processes, including long-term potentiation. Alterations in mechanisms related to ERK are associated with different pathological outcomes. Genetic alterations in any component of the ERK pathway result in pathologies associated with neural crest derivatives and mental dysfunctions associated with autism spectrum disorders. The MAP-ERK pathway is a key element of the neuroinflammatory pathway triggered by glial cells during the development of neurodegenerative diseases, such as Parkinson's and Alzheimer's disease, Huntington's disease, and amyotrophic lateral sclerosis, as well as prionic diseases. The process triggered by MAPK/ERK activation depends on the stage of development (mature or senescence), the type of cellular element in which the pathway is activated, and the anatomic neural structure. However, extensive gaps exist with regards to the targets of the phosphorylated ERK in many of these processes.
Collapse
Affiliation(s)
- Héctor Albert-Gascó
- UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Hills Road, Cambridge CB2 0AH, UK;
| | - Francisco Ros-Bernal
- U.P Medicina, Facultad de Ciencias de la Salud, Universitat Jaume I, Avda. de Vicent Sos Baynat s/n, 12071 Castelló de la Plana, Spain; (F.R.-B.); (E.C.-G.)
| | - Esther Castillo-Gómez
- U.P Medicina, Facultad de Ciencias de la Salud, Universitat Jaume I, Avda. de Vicent Sos Baynat s/n, 12071 Castelló de la Plana, Spain; (F.R.-B.); (E.C.-G.)
- Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Planta 0, 28029 Madrid, Spain
| | - Francisco E. Olucha-Bordonau
- U.P Medicina, Facultad de Ciencias de la Salud, Universitat Jaume I, Avda. de Vicent Sos Baynat s/n, 12071 Castelló de la Plana, Spain; (F.R.-B.); (E.C.-G.)
- Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Planta 0, 28029 Madrid, Spain
| |
Collapse
|
22
|
Harricharran T, Ogunwobi OO. Oxytocin and oxytocin receptor alterations, decreased survival, and increased chemoresistance in patients with pancreatic cancer. Hepatobiliary Pancreat Dis Int 2020; 19:175-180. [PMID: 31919036 PMCID: PMC7265130 DOI: 10.1016/j.hbpd.2019.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 12/04/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND Oxytocin (OXT) and its receptor (OXTR) is associated with cancer. The present study was to investigate the correlation between the genetic expression alterations of OXT and OXTR and the outcomes in patients with pancreatic cancer (PC). METHODS Information regarding OXT and OXTR genetic alterations and changes in gene expression were retrieved from the Cancer Genome Atlas (TCGA) databases and analyzed using the cBioPortal online tool. We assessed the correlation of overall survival and disease/progression-free months to either OXT or OXTR genetic alterations and changes in gene expression using Kaplan-Meier and Cox regression analyses. Quantitative PCR (qPCR) was conducted to assess the mRNA expression levels of OXT and OXTR in human PC cell lines. RESULTS Five percent of PC cases showed mRNA upregulation in the OXT gene. These PC cases also showed genetic alterations and changes in gene expression of OXTR. The median months of survival and disease-free survival were lower for PC cases with genetic alterations and changes in gene expression in the OXT and OXTR genes as compared to those without such alterations. qPCR data showed that OXT and OXTR mRNA expression were 1-fold and 10-fold higher, respectively in PANC-1 cell lines as compared to L3.6pl cell lines in direct negative correlation with responsiveness to gemcitabine. CONCLUSIONS These data suggest that OXT and OXTR may potentially be important in PC progression, chemoresistance, and patient survival, and potentially could have prognostic and therapeutic implications in a subset of PC patients.
Collapse
Affiliation(s)
- Trisheena Harricharran
- Department of Biological Sciences, Hunter College of The City University of New York, New York, NY 10065, USA; The Graduate Center Departments of Biology and Biochemistry, The City University of New York, New York, NY 10016, USA; Hunter College Center for Cancer Health Disparities Research (CCHDR), New York, NY 10065, USA
| | - Olorunseun O Ogunwobi
- Department of Biological Sciences, Hunter College of The City University of New York, New York, NY 10065, USA; The Graduate Center Departments of Biology and Biochemistry, The City University of New York, New York, NY 10016, USA; Hunter College Center for Cancer Health Disparities Research (CCHDR), New York, NY 10065, USA; Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA.
| |
Collapse
|
23
|
Myocyte Enhancer Factor 2A (MEF2A) Defines Oxytocin-Induced Morphological Effects and Regulates Mitochondrial Function in Neurons. Int J Mol Sci 2020; 21:ijms21062200. [PMID: 32209973 PMCID: PMC7139413 DOI: 10.3390/ijms21062200] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/16/2020] [Accepted: 03/20/2020] [Indexed: 02/06/2023] Open
Abstract
The neuropeptide oxytocin (OT) is a well-described modulator of socio-emotional traits, such as anxiety, stress, social behavior, and pair bonding. However, when dysregulated, it is associated with adverse psychiatric traits, such as various aspects of autism spectrum disorder (ASD). In this study, we identify the transcription factor myocyte enhancer factor 2A (MEF2A) as the common link between OT and cellular changes symptomatic for ASD, encompassing neuronal morphology, connectivity, and mitochondrial function. We provide evidence for MEF2A as the decisive factor defining the cellular response to OT: while OT induces neurite retraction in MEF2A expressing neurons, OT causes neurite outgrowth in absence of MEF2A. A CRISPR-Cas-mediated knockout of MEF2A and retransfection of an active version or permanently inactive mutant, respectively, validated our findings. We also identified the phosphatase calcineurin as the main upstream regulator of OT-induced MEF2A signaling. Further, MEF2A signaling dampens mitochondrial functioning in neurons, as MEF2A knockout cells show increased maximal cellular respiration, spare respiratory capacity, and total cellular ATP. In summary, we reveal a central role for OT-induced MEF2A activity as major regulator of cellular morphology as well as neuronal connectivity and mitochondrial functioning, with broad implications for a potential treatment of disorders based on morphological alterations or mitochondrial dysfunction.
Collapse
|
24
|
The Role of the Oxytocin System in Anxiety Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1191:103-120. [PMID: 32002925 DOI: 10.1007/978-981-32-9705-0_7] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Oxytocin, a neuropeptide synthesized by the hypothalamus, plays a central role in human social behavior, social cognition, anxiety, mood, stress modulation, and fear learning and extinction. The relationships between oxytocin and psychiatric disorders including depression, anxiety, schizophrenia, and autism spectrum disorder have been extensively studied. In this chapter, we focus on the current knowledge about oxytocin and anxiety disorder. We discuss the anxiolytic effects of oxytocin in preclinical and clinical findings, possible related neurobehavioral mechanisms (social cognition, fear learning, and extinction), related neurotransmitter and neuroendocrine systems (hypothalamus-pituitary-adrenal axis, serotoninergic, and GABAergic systems), and studies regarding plasma levels of oxytocin, genetic and epigenetic findings, and effects of intranasal oxytocin in DSM-5 anxiety disorder (primarily social anxiety disorder and separation anxiety disorder) patients.
Collapse
|
25
|
Jurek B, Meyer M. Anxiolytic and Anxiogenic? How the Transcription Factor MEF2 Might Explain the Manifold Behavioral Effects of Oxytocin. Front Endocrinol (Lausanne) 2020; 11:186. [PMID: 32322239 PMCID: PMC7156634 DOI: 10.3389/fendo.2020.00186] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/17/2020] [Indexed: 01/01/2023] Open
Abstract
The neuromodulator oxytocin, since its first synthesis by du Vigneaud in 1953, has mainly been associated with beneficial physiological effects, as well as positive social and emotional behaviors. This overall positive picture of oxytocin as the "love-, cuddle-, or bonding-hormone" has repeatedly been challenged since then. Oxytocin-induced effects that would be perceived as negative by the individual, such as increased anxiety or potentiation of stress-induced ACTH release, as well as the regulation of negative approach-related emotions, such as envy and schadenfreude (gloating) have been described. The general consent is that oxytocin, instead of acting unidirectional, induces changes in the salience network to shift the emphasis of emotional contexts, and therefore can, e.g., produce both anxiolytic as well as anxiogenic behavioral outcomes. However, the underlying mechanisms leading to alterations in the salience network are still unclear. With the aim to understand the manifold effects of oxytocin on a cellular/molecular level, a set of oxytocin receptor-coupled signaling cascades and downstream effectors regulating transcription and translation has been identified. Those oxytocin-driven effectors, such as MEF2 and CREB, are known modulators of the neuronal and glial cytoarchitecture. We hypothesize that, by determining cellular morphology and connectivity, MEF2 is one of the key factors that might contribute to the diverse behavioral effects of oxytocin.
Collapse
|
26
|
Martinetz S, Meinung CP, Jurek B, von Schack D, van den Burg EH, Slattery DA, Neumann ID. De Novo Protein Synthesis Mediated by the Eukaryotic Elongation Factor 2 Is Required for the Anxiolytic Effect of Oxytocin. Biol Psychiatry 2019; 85:802-811. [PMID: 30826070 DOI: 10.1016/j.biopsych.2019.01.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 12/17/2018] [Accepted: 01/02/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND The neuropeptide oxytocin (OXT) mediates its actions, including anxiolysis, via its G protein-coupled OXT receptor. Within the paraventricular nucleus of the hypothalamus (PVN), OXT-induced anxiolysis is mediated, at least in part, via activation of the mitogen-activated protein kinase pathway following calcium influx through transient receptor potential cation channel subfamily V member 2 channels. In the periphery, OXT activates eukaryotic elongation factor 2 (eEF2), an essential mediator of protein synthesis. METHODS In order to study whether OXT activates eEF2 also in neurons to exert its anxiolytic properties in the PVN, we performed in vivo and cell culture experiments. RESULTS We demonstrate that OXT, in a protein kinase C-dependent manner, activates eEF2 both in a hypothalamic cell line and in vivo within the PVN. Next, we reveal that OXT stimulates de novo protein synthesis, while inhibition of protein synthesis within the PVN prevents the anxiolytic effect of OXT in male rats. Moreover, activation of eEF2 within the PVN conveyed an anxiolytic effect supporting a role of OXT-induced eEF2 activation and protein synthesis for its anxiolysis. Finally, we show that one of the proteins that is upregulated by OXT is the neuropeptide Y receptor 5. Infusion of a specific neuropeptide Y receptor 5 agonist into the PVN consequently led to decreased anxiety-related behavior, while pretreatment with a neuropeptide Y receptor 5 antagonist prevented the anxiolytic effect of OXT. CONCLUSIONS Taken together, these results show that OXT recruits several intracellular signaling cascades to induce protein synthesis, which mediates the anxiolytic effects of OXT within the PVN and suggests that eEF2 represents a novel target for anxiety-related disorders.
Collapse
Affiliation(s)
- Stefanie Martinetz
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Carl-Philipp Meinung
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - Benjamin Jurek
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| | - David von Schack
- Biotherapeutics Clinical Research and Development, Precision Medicine, New York, New York
| | | | - David A Slattery
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany; Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Inga D Neumann
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
27
|
Zatkova M, Reichova A, Bacova Z, Bakos J. Activation of the Oxytocin Receptor Modulates the Expression of Synaptic Adhesion Molecules in a Cell-Specific Manner. J Mol Neurosci 2019; 68:171-180. [DOI: 10.1007/s12031-019-01296-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 03/11/2019] [Indexed: 11/29/2022]
|
28
|
Neumann ID, Landgraf R. Tracking oxytocin functions in the rodent brain during the last 30 years: From push-pull perfusion to chemogenetic silencing. J Neuroendocrinol 2019; 31:e12695. [PMID: 30748037 DOI: 10.1111/jne.12695] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 12/22/2022]
Abstract
A short overview is provided of the last 30 years of oxytocin (and vasopressin) research performed in our laboratories, starting with attempts to monitor the release of this nonapeptide in the rodent brain during physiological conditions such as suckling in the lactating animal. Using push-pull perfusion and microdialysis approaches, release patterns in hypothalamic and limbic brain regions could be characterised to occur from intact neuronal structures, to be independent of peripheral secretion into blood, and to respond differentially to various stimuli, particularly those related to reproduction and stress. Parallel efforts focused on the functional impact of central oxytocin release, including neuroendocrine and behavioural effects mediated by nonapeptide receptor interactions and subsequent intraneuronal signalling cascades. The use of a variety of sophisticated behavioural paradigms to manipulate central oxytocin release, along with pharmacological, genetic and pharmacogenetic approaches, revealed multiple consequences on social behaviours, particularly social fear.
Collapse
Affiliation(s)
- Inga D Neumann
- Department of Behavioural and Molecular Neurobiology, Regensburg Centre of Neurosciences, University of Regensburg, Regensburg, Germany
| | | |
Collapse
|
29
|
Targeting the Oxytocin System: New Pharmacotherapeutic Approaches. Trends Pharmacol Sci 2019; 40:22-37. [DOI: 10.1016/j.tips.2018.11.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/27/2018] [Accepted: 11/01/2018] [Indexed: 12/27/2022]
|
30
|
Acute and long-lasting effects of oxytocin in cortico-limbic circuits: consequences for fear recall and extinction. Psychopharmacology (Berl) 2019; 236:339-354. [PMID: 30302511 DOI: 10.1007/s00213-018-5030-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 09/05/2018] [Indexed: 12/11/2022]
Abstract
The extinction of conditioned fear responses entrains the formation of safe new memories to decrease those behavioral responses. The knowledge in neuronal mechanisms of extinction is fundamental in the treatment of anxiety and fear disorders. Interestingly, the use of pharmacological compounds that reduce anxiety and fear has been shown as a potent co-adjuvant in extinction therapy. However, the efficiency and mechanisms by which pharmacological compounds promote extinction of fear memories remains still largely unknown and would benefit from a validation based on functional neuronal circuits, and the neurotransmitters that modulate them. From this perspective, oxytocin receptor signaling, which has been shown in cortical and limbic areas to modulate numerous functions (Eliava et al. Neuron 89(6):1291-1304, 2016), among them fear and anxiety circuits, and to enhance the salience of social stimuli (Stoop Neuron 76(1):142-59, 2012), may offer an interesting perspective. Experiments in animals and humans suggest that oxytocin could be a promising pharmacological agent at adjusting memory consolidation to boost fear extinction. Additionally, it is possible that long-term changes in endogenous oxytocin signaling can also play a role in reducing expression of fear at different brain targets. In this review, we summarize the effects reported for oxytocin in cortico-limbic circuits and on fear behavior that are of relevance for the modulation and potential extinction of fear memories.
Collapse
|
31
|
Meyer M, Berger I, Winter J, Jurek B. Oxytocin alters the morphology of hypothalamic neurons via the transcription factor myocyte enhancer factor 2A (MEF-2A). Mol Cell Endocrinol 2018; 477:156-162. [PMID: 29928931 DOI: 10.1016/j.mce.2018.06.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/18/2018] [Accepted: 06/16/2018] [Indexed: 01/22/2023]
Abstract
Oxytocin (OT) has gained attention not only as anxiolytic drug and as potential treatment option for autistic children; it also acts as a growth and differentiation factor in neuronal cells. While behavioral effects of OT have been studied in detail, knowledge about the cellular effects of OT is relatively sparse. In this study, we present evidence for three hypotheses: 1) OT leads to neurite retraction in hypothalamic neurons via the OT receptor (OTR) 2) The transcription factor MEF-2A is a central regulator of OT-induced neurite retraction, and 3) The MAPK pathway is critical for OT-induced MEF-2A activation. Incubation of rat hypothalamic H32 cells with 10 nM to 1 μM OT, vasopressin, and the specific OTR agonist TGOT, over the course of 12 h resulted in a time-dependent, significant retraction of neurites. In addition, the size of the nuclear compartment increased, whereas the overall cell size remained unchanged. OT treatment for 10 h increased the cellular viability significantly, and this effect could be blocked by a specific OTR antagonist, providing evidence for a specific and pro-active effect of OT on neurite retraction, and not as an unspecific side effect of apoptosis. The molecular mechanism that controls OT-induced neurite retraction includes a reduced phosphorylation of the transcription factor MEF-2A at Serine 408 (S408). This dephosphorylation is under the control of the OTR-coupled MAPK pathway, as blocking MEK1/2 by U0126 inhibited MEF-2A activation and subsequent neurite retraction. The siRNA-mediated knockdown of MEF-2A prevented the OT-induced neurite retraction, providing direct evidence for a role of MEF-2A in morphological alterations induced by OT treatment. In summary, the present study reveals a previously unknown OTR-coupled MAPK-MEF-2A pathway, which is responsible for OT-induced neurite retraction of hypothalamic neurons.
Collapse
Affiliation(s)
- Magdalena Meyer
- Department of Behavioral and Molecular Neurobiology, University of Regensburg, Regensburg, Germany.
| | - Ilona Berger
- Department of Behavioral and Molecular Neurobiology, University of Regensburg, Regensburg, Germany; Technische Universität Dresden, University Hospital, Department of Internal Medicine III, Dresden, Germany
| | - Julia Winter
- Department of Behavioral and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| | - Benjamin Jurek
- Department of Behavioral and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
32
|
Mustoe A, Taylor JH, French JA. Oxytocin structure and function in New World monkeys: from pharmacology to behavior. Integr Zool 2018; 13:634-654. [PMID: 29436774 PMCID: PMC6089668 DOI: 10.1111/1749-4877.12318] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Oxytocin (OT) is a hypothalamic nonapeptide that mediates a host of physiological and behavioral processes including reproductive physiology and social attachments. While the OT sequence structure is highly conserved among mammals, New World monkeys (NWMs) represent an unusual "hot spot" in OT structure variability among mammals. At least 6 distinct OT ligand variants among NWMs exist, yet it is currently unclear whether these evolved structural changes result in meaningful functional consequences. NWMs offer a new area to explore how these modifications to OT and its canonical G-protein coupled OT receptor (OTR) may mediate specific cellular, physiological and behavioral outcomes. In this review, we highlight relationships between OT ligand and OTR structural variability, specifically examining coevolution between OT ligands, OTRs, and physiological and behavioral phenotypes across NWMs. We consider whether these evolved modifications to the OT structure alter pharmacological profiles at human and marmoset OTRs, including changes to receptor binding, intracellular signaling and receptor internalization. Finally, we evaluate whether exogenous manipulation using OT variants in marmoset monkeys differentially enhance or impair behavioral processes involved in social relationships between pairmates, opposite-sex strangers, and parents and their offspring. Overall, it appears that changes to OT ligands in NWMs result in important changes ranging from cellular signaling to broad measures of social behavior.
Collapse
Affiliation(s)
- Aaryn Mustoe
- Program in Neuroscience and Behavior, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Jack H Taylor
- Program in Neuroscience and Behavior, University of Nebraska at Omaha, Omaha, Nebraska, USA
| | - Jeffrey A French
- Program in Neuroscience and Behavior, University of Nebraska at Omaha, Omaha, Nebraska, USA
| |
Collapse
|
33
|
Yang B, Jiao B, Ge W, Zhang X, Wang S, Zhao H, Wang X. Transcriptome sequencing to detect the potential role of long non-coding RNAs in bovine mammary gland during the dry and lactation period. BMC Genomics 2018; 19:605. [PMID: 30103699 PMCID: PMC6090732 DOI: 10.1186/s12864-018-4974-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 07/31/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND It is known that long non-coding RNAs (lncRNAs) play an important role in various biological processes, including cell proliferation, differentiation and apoptosis. However, their functions and profiles in lactation cycle of dairy cows are largely unknown. In this study, lncRNA-seq technique was employed to compare the expression profiles of lncRNAs and mRNAs from Chinese Holstein mammary gland in dry and lactation period. RESULT Totally 3746 differentially expressed lncRNAs (DELs) and 2890 differentially expressed genes (DEGs) were identified from the dry and lactation mammary glands of Holstein cows. Functional enrichment analysis on target genes of lncRNAs indicated that these genes were involved in lactation-related signaling pathways, including cell cycle, JAK-STAT, cell adhesion, and PI3K-Akt signaling pathways. Additionally, the interaction between lncRNAs and their potential miRNAs was predicted and partly verified. The result indicated that the lactation-associated miR-221 might interact with lncRNAs TCONS_00040268, TCONS_00137654, TCONS_00071659 and TCONS_00000352, which revealed that these lncRNAs might be important regulators for lactation cycle. CONCLUSION This study provides a resource for lncRNA research on lactation cycle of bovine mammary gland. Besides, the interaction between lncRNAs and the specific miRNA is revealed. It expands our knowledge about lncRNA and miRNA biology as well as contributes to clarify the regulation of lactation cycle of bovine mammary gland.
Collapse
Affiliation(s)
- Bing Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China.,Lab of Feed and Animal Nutrition, Tongren Polytechnic College, Tongren, 554300, Guizhou, China
| | - Beilei Jiao
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Wei Ge
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Xiaolan Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Shanhe Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Hongbo Zhao
- Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, Shandong, China
| | - Xin Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
34
|
HDAC9 promotes brain ischemic injury by provoking IκBα/NF-κB and MAPKs signaling pathways. Biochem Biophys Res Commun 2018; 503:1322-1329. [PMID: 30031609 DOI: 10.1016/j.bbrc.2018.07.043] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 07/09/2018] [Indexed: 12/17/2022]
Abstract
Ischemic stroke is an acute cerebrovascular disease due to poor blood flow to the brain. Nevertheless, there is still no effective therapy for it and the pathology contributing to ischemic stroke is not fully understood. Histone Deacetylase 9 (HDAC9) is a class IIa chromatin-modifying enzyme. HDAC9 gene region is a leading risk locus for large artery atherosclerotic stroke. However, the mechanisms linking HDAC9 to ischemic remain elusive. In the study, we attempted to explore HDAC9-associated inflammatory response using the wild type (WT) and HDAC9-knockout (KO) mice with brain ischemic injury. The results indicated that WT mice with ischemia brain exhibited higher expression levels of HDAC9. HDAC9 depletion resulted in a decreased infarct volume and an improved neurological function in mice after ischemic reperfusion (I/R) injury. I/R injury markedly enhanced GFAP and Iba-1 expressions in cortex and HDAC9 knockout significantly reversed this up-regulation. Loss of HDAC9 inhibited the release of inducible NO-synthase (iNOS), cyclooxygenase-2 (COX-2), interleukin 1β (IL-1β), IL-6, tumor necrosis factor-α (TNF-α), and IL-18 in cortex, hippocampus and hypothalamus of mice with I/R injury, which occurred at the transcription levels. Furthermore, the inhibitory actions of HDAC9 deficiency were associated with the down-regulation of phosphorylated-IκBα, phosphorylated-nuclear factor-kappa B (NF-κB), and p-mitogen-activated protein kinases (MAPKs), including phosphorylated-p38, phosphorylated-extracellular signal-regulated kinase 1/2 (ERK1/2), and phosphorylated-c-Jun N-terminal kinase (JNK). Importantly, the in vitro study indicated that HDAC9 inhibition-reduced inflammation and activation of IκBα/NF-κB were restored by promoting MAPKs activity in LPS-stimulated cells. Our findings suggest that HDAC9 inhibition showed neuroprotective effects on ischemic stroke by restraining inflammation, which might help develop new and effective strategies for the therapeutic interventions in ischemic stroke.
Collapse
|
35
|
Sun W, Zhou Q, Ba X, Feng X, Hu X, Cheng X, Liu T, Guo J, Xiao L, Jiang J, Xiong D, Hao Y, Chen Z, Jiang C. Oxytocin Relieves Neuropathic Pain Through GABA Release and Presynaptic TRPV1 Inhibition in Spinal Cord. Front Mol Neurosci 2018; 11:248. [PMID: 30065629 PMCID: PMC6056657 DOI: 10.3389/fnmol.2018.00248] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/28/2018] [Indexed: 01/14/2023] Open
Abstract
Objective: Oxytocin (OT) is synthesized within the paraventricular nucleus and supraoptic nucleus of the hypothalamus. In addition to its role in uterine contraction, OT plays an important antinociceptive role; however, the underlying molecular mechanisms of antinociceptive role of OT remain elusive. We hypothesized that the antinociceptive effect of OT on neuropathic pain may occur via inhibition of TRPV1 activation in the spinal cord. The present study explores the antinociceptive role of OT and its mechanisms in neuropathic pain. Methods: Partial sciatic nerve ligation (pSNL) was performed to induce neuropathic pain. Animal behaviors were measured using a set of electronic von Frey apparatus and hot plate. Electrophysiological recordings and molecular biological experiments were performed. Results: Intrathecal administration of OT alleviated both mechanical allodynia and thermal hyperalgesia in pSNL rats (n = 6, per group, P < 0.0001, saline vs. OT group). Electrophysiological data revealed that OT significantly inhibited the enhancement of frequency and amplitude of spontaneous excitatory post-synaptic currents induced presynaptically by TRPV1 activation in the spinal cord. Moreover, the inhibitory effect of OT on capsaicin-induced facilitation of excitatory transmission was blocked by co-treatment with saclofen, while intrathecal administration of OT dramatically inhibited capsaicin-induced ongoing pain in rats, (n = 6, per group, P < 0.0001, saline vs. OT group). The paw withdrawal latency in response to heat stimulation was significantly impaired in TRPV1KO mice 3 days after pSNL upon OT (i.t.) treatment, compared with wild type mice (n = 6, P < 0.05). Finally, OT prevented TRPV1 up-regulation in spinal cords of pSNL model rats. Conclusion: OT relieves neuropathic pain through GABA release and presynaptic TRPV1 inhibition in the spinal cord. OT and its receptor system might be an intriguing target for the treatment and prevention of neuropathic pain.
Collapse
Affiliation(s)
- Wuping Sun
- Department of Pain Medicine, Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Qian Zhou
- Department of Pain Medicine, The Third People's Hospital of Hubei Province, Wuhan, China
| | - Xiyuan Ba
- Department of Pain Medicine, Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Xiaojin Feng
- Center for Experimental Medicine, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Xuexue Hu
- Center for Experimental Medicine, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Xiaoe Cheng
- Center for Experimental Medicine, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Tao Liu
- Center for Experimental Medicine, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Jing Guo
- Department of Pain Medicine, Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Lizu Xiao
- Department of Pain Medicine, Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Jin Jiang
- Department of Pain Medicine, Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Donglin Xiong
- Department of Pain Medicine, Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| | - Yue Hao
- Department of Pharmacy, School of Medicine, Health Science Center, Shenzhen University, Shenzhen, China
| | - Zixian Chen
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Changyu Jiang
- Department of Pain Medicine, Shenzhen Municipal Key Laboratory for Pain Medicine, Shenzhen Nanshan People's Hospital and the 6th Affiliated Hospital, Health Science Center, Shenzhen University, Shenzhen, China
| |
Collapse
|
36
|
Jurek B, Neumann ID. The Oxytocin Receptor: From Intracellular Signaling to Behavior. Physiol Rev 2018; 98:1805-1908. [DOI: 10.1152/physrev.00031.2017] [Citation(s) in RCA: 601] [Impact Index Per Article: 85.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The many facets of the oxytocin (OXT) system of the brain and periphery elicited nearly 25,000 publications since 1930 (see FIGURE 1 , as listed in PubMed), which revealed central roles for OXT and its receptor (OXTR) in reproduction, and social and emotional behaviors in animal and human studies focusing on mental and physical health and disease. In this review, we discuss the mechanisms of OXT expression and release, expression and binding of the OXTR in brain and periphery, OXTR-coupled signaling cascades, and their involvement in behavioral outcomes to assemble a comprehensive picture of the central and peripheral OXT system. Traditionally known for its role in milk let-down and uterine contraction during labor, OXT also has implications in physiological, and also behavioral, aspects of reproduction, such as sexual and maternal behaviors and pair bonding, but also anxiety, trust, sociability, food intake, or even drug abuse. The many facets of OXT are, on a molecular basis, brought about by a single receptor. The OXTR, a 7-transmembrane G protein-coupled receptor capable of binding to either Gαior Gαqproteins, activates a set of signaling cascades, such as the MAPK, PKC, PLC, or CaMK pathways, which converge on transcription factors like CREB or MEF-2. The cellular response to OXT includes regulation of neurite outgrowth, cellular viability, and increased survival. OXTergic projections in the brain represent anxiety and stress-regulating circuits connecting the paraventricular nucleus of the hypothalamus, amygdala, bed nucleus of the stria terminalis, or the medial prefrontal cortex. Which OXT-induced patterns finally alter the behavior of an animal or a human being is still poorly understood, and studying those OXTR-coupled signaling cascades is one initial step toward a better understanding of the molecular background of those behavioral effects.
Collapse
Affiliation(s)
- Benjamin Jurek
- Department of Behavioural and Molecular Neurobiology, Institute of Zoology, University of Regensburg, Regensburg, Germany
| | - Inga D. Neumann
- Department of Behavioural and Molecular Neurobiology, Institute of Zoology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
37
|
Rajamani KT, Wagner S, Grinevich V, Harony-Nicolas H. Oxytocin as a Modulator of Synaptic Plasticity: Implications for Neurodevelopmental Disorders. Front Synaptic Neurosci 2018; 10:17. [PMID: 29970997 PMCID: PMC6018411 DOI: 10.3389/fnsyn.2018.00017] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 05/25/2018] [Indexed: 01/23/2023] Open
Abstract
The neuropeptide oxytocin (OXT) is a crucial mediator of parturition and milk ejection and a major modulator of various social behaviors, including social recognition, aggression and parenting. In the past decade, there has been significant excitement around the possible use of OXT to treat behavioral deficits in neurodevelopmental disorders, including autism spectrum disorder (ASD). Yet, despite the fast move to clinical trials with OXT, little attention has been paid to the possibility that the OXT system in the brain is perturbed in these disorders and to what extent such perturbations may contribute to social behavior deficits. Large-scale whole-exome sequencing studies in subjects with ASD, along with biochemical and electrophysiological studies in animal models of the disorder, indicate several risk genes that play an essential role in brain synapses, suggesting that deficits in synaptic activity and plasticity underlie the pathophysiology in a considerable portion of these cases. OXT has been repeatedly shown, both in vitro and in vivo, to modify synaptic properties and plasticity and to modulate neural activity in circuits that regulate social behavior. Together, these findings led us to hypothesize that failure of the OXT system during early development, as a direct or indirect consequence of genetic mutations, may impact social behavior by altering synaptic activity and plasticity. In this article, we review the evidence that support our hypothesis.
Collapse
Affiliation(s)
- Keerthi Thirtamara Rajamani
- The Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,The Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Shlomo Wagner
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Valery Grinevich
- Schaller Research Group on Neuropeptides at German Cancer Research Center (DKFZ), Central Institute of Mental Health and Cell Networks Cluster of Excellence, University of Heidelberg, Heidelberg, Germany
| | - Hala Harony-Nicolas
- The Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, United States.,The Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| |
Collapse
|
38
|
Oxytocin Signaling in the Lateral Septum Prevents Social Fear during Lactation. Curr Biol 2018; 28:1066-1078.e6. [DOI: 10.1016/j.cub.2018.02.044] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/09/2018] [Accepted: 02/16/2018] [Indexed: 01/03/2023]
|
39
|
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
|
40
|
Grund T, Goyon S, Li Y, Eliava M, Liu H, Charlet A, Grinevich V, Neumann ID. Neuropeptide S Activates Paraventricular Oxytocin Neurons to Induce Anxiolysis. J Neurosci 2017; 37:12214-12225. [PMID: 29118105 PMCID: PMC6596824 DOI: 10.1523/jneurosci.2161-17.2017] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/27/2017] [Accepted: 10/31/2017] [Indexed: 01/12/2023] Open
Abstract
Neuropeptides, such as neuropeptide S (NPS) and oxytocin (OXT), represent potential options for the treatment of anxiety disorders due to their potent anxiolytic profile. In this study, we aimed to reveal the mechanisms underlying the behavioral action of NPS, and present a chain of evidence that the effects of NPS within the hypothalamic paraventricular nucleus (PVN) are mediated via actions on local OXT neurons in male Wistar rats. First, retrograde studies identified NPS fibers originating in the brainstem locus coeruleus, and projecting to the PVN. FACS identified prominent NPS receptor expression in PVN-OXT neurons. Using genetically encoded calcium indicators, we further demonstrated that NPS reliably induces a transient increase in intracellular Ca2+ concentration in a subpopulation of OXT neurons, an effect mediated by NPS receptor. In addition, intracerebroventricular (i.c.v.) NPS evoked a significant somatodendritic release of OXT within the PVN as assessed by microdialysis in combination with a highly sensitive radioimmunoassay. Finally, we could show that the anxiolytic effect of NPS seen after i.c.v. or intra-PVN infusion requires responsive OXT neurons of the PVN and locally released OXT. Thus, pharmacological blockade of OXT receptors as well as chemogenetic silencing of OXT neurons within the PVN prevented the effect of synthetic NPS. In conclusion, our results indicate a significant role of the OXT system in mediating the effects of NPS on anxiety, and fill an important gap in our understanding of brain neuropeptide interactions in the context of regulation of emotional behavior within the hypothalamus.SIGNIFICANCE STATEMENT Given the rising scientific interest in neuropeptide research in the context of emotional and stress-related behaviors, our findings demonstrate a novel intrahypothalamic mechanism involving paraventricular oxytocin neurons that express the neuropeptide S receptor. These neurons respond with transient Ca2+ increase and somatodendritic oxytocin release following neuropeptide S stimulation. Thereby, oxytocin neurons seem essential for neuropeptide S-induced anxiolysis, as this effect was blocked by pharmacological and chemogenetic inhibition of the oxytocin system.
Collapse
Affiliation(s)
- Thomas Grund
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, 93040 Regensburg, Germany
| | - Stephanie Goyon
- Institute of Cellular and Integrative Neurosciences UPR3212, Centre National de la Recherche Scientifique, University of Strasbourg, 67084 Strasbourg, France
| | - Yuting Li
- Division of Molecular Neurogenetics, German Cancer Research Center, DKFZ-ZMBH Alliance, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Marina Eliava
- Division of Neuropeptides (V078), German Cancer Research Center, Central Institute of Mental Health, CellNetwork Cluster of Excellence, University of Heidelberg, 69120 Heidelberg, Germany, and
| | - Haikun Liu
- Division of Molecular Neurogenetics, German Cancer Research Center, DKFZ-ZMBH Alliance, Im Neuenheimer Feld 280, Heidelberg 69120, Germany
| | - Alexandre Charlet
- Institute of Cellular and Integrative Neurosciences UPR3212, Centre National de la Recherche Scientifique, University of Strasbourg, 67084 Strasbourg, France
- University of Strasbourg, Institute for Advanced Study, 67084 Strasbourg, France
| | - Valery Grinevich
- Division of Neuropeptides (V078), German Cancer Research Center, Central Institute of Mental Health, CellNetwork Cluster of Excellence, University of Heidelberg, 69120 Heidelberg, Germany, and
| | - Inga D Neumann
- Department of Behavioral and Molecular Neurobiology, Regensburg Center of Neuroscience, University of Regensburg, 93040 Regensburg, Germany,
| |
Collapse
|
41
|
Dong N, Du P, Hao X, He Z, Hou W, Wang L, Yuan W, Yang J, Jia R, Tai F. Involvement of GABA A receptors in the regulation of social preference and emotional behaviors by oxytocin in the central amygdala of female mandarin voles. Neuropeptides 2017; 66:8-17. [PMID: 28764883 DOI: 10.1016/j.npep.2017.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 05/19/2017] [Accepted: 07/24/2017] [Indexed: 12/31/2022]
Abstract
The central nucleus of the amygdala (CeA) is the main output of the amygdala and plays an important role in behavioral and neuroendocrine responses to stress. Receptors for the neuropeptide oxytocin (OT) and GABAA are found in high concentration in the CeA. The mechanisms underlying regulation of CeA OT in emotional and social behavior remain unclear. In this study we evaluated the effects of intra-CeA OT administration of different doses (0.1, 1 and 10ng/side), OT receptor antagonist (OTR-A) (1, 10 and 100ng/side) and OT plus OTR-A on social and emotional behavior using a social preference paradigm, open field test and elevated plus maze test in female monogamous mandarin voles (Microtus mandarinus). We then examined whether different doses of the GABAA receptor antagonist bicuculline (5, 10 and 100ng/side) affected the behavioral changes induced by intra-CeA microinjection of OT (1ng/side). We found that administration of OT to the CeA increased social preference, central area investigation times in the open field test, and visits, transitions and time spent in the open arms in the elevated plus maze test; all responses were dose-dependent. Administration of OT plus OTR-A to the CeA produced no effects. Administration of bicuculline in combination with OT to the CeA decreased social preference, central area investigation times in the open field test, and visits, transitions and time spent in the open arms of the elevated plus maze test. These data suggest that OT in the CeA facilitates sociality and reduces levels of anxiety by interacting with local GABAA receptors.
Collapse
Affiliation(s)
- Na Dong
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710100, China
| | - Peirong Du
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710100, China
| | - Xin Hao
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710100, China
| | - Zhixiong He
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710100, China
| | - Wenjuan Hou
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710100, China
| | - Limin Wang
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710100, China
| | - Wei Yuan
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710100, China
| | - Jinfeng Yang
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710100, China
| | - Rui Jia
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710100, China.
| | - Fadao Tai
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710100, China.
| |
Collapse
|
42
|
Ong ZY, Bongiorno DM, Hernando MA, Grill HJ. Effects of Endogenous Oxytocin Receptor Signaling in Nucleus Tractus Solitarius on Satiation-Mediated Feeding and Thermogenic Control in Male Rats. Endocrinology 2017; 158:2826-2836. [PMID: 28575174 PMCID: PMC5659667 DOI: 10.1210/en.2017-00200] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 05/24/2017] [Indexed: 12/15/2022]
Abstract
Central oxytocin receptor (OT-R) signaling reduces food intake and increases energy expenditure, but the central sites and mechanisms mediating these effects are unresolved. We showed previously that pharmacological activation of OT-R in hindbrain/nucleus tractus solitarius (NTS) amplifies the intake-inhibitory effects of gastrointestinal (GI) satiation signals. Unexplored were the energetic effects of hindbrain OT-R agonism and the physiological relevance of NTS OT-R signaling on food intake and energy expenditure control. Using a virally mediated OT-R knockdown (KD) strategy and a range of behavioral paradigms, this study examined the role of endogenous NTS OT-R signaling on satiation-mediated food intake inhibition and thermogenic control. Results showed that, compared with controls, NTS OT-R KD rats consumed larger meals, were less responsive to the intake-inhibitory effects of a self-ingested preload, and consumed more chow following a 24-hour fast. These data indicate that NTS OT-R signaling is necessary for normal satiation control. Whereas both control and NTS OT-R KD rats increased core temperature following high-fat diet maintenance (relative to chow maintenance), the percent increase in core temperature was greater in control compared with NTS OT-R KD rats during the light cycle. Hindbrain oxytocin agonist delivery increased core temperature in both control and NTS OT-R KD rats and the percent increase relative to vehicle treatment was not significantly different between groups. Together, data reveal a critical role for endogenous NTS OT-R signaling in mediating the intake-inhibitory effects of endogenous GI satiation signals and in diet-induced thermogenesis.
Collapse
Affiliation(s)
- Zhi Yi Ong
- Department of Psychology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Diana M. Bongiorno
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Mary Ann Hernando
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Harvey J. Grill
- Department of Psychology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW This review aims to evaluate the most recent literature examining the oxytocin (OXT) system's role in human anxiety by surveying various fields of preclinical and clinical research supporting this role, and queries whether the OXT system might be a target for novel anxiolytics. RECENT FINDINGS Evidence from the diverse body of literature presented here, from translational research, genetic and neuroimaging studies, to clinical trials of intranasal (IN) OXT reveals a positive association. In addition, some moderators (e.g., sex, specificities to cues) of OXT's anxiolytic effects can have an important influence on its outcomes, awaiting further research. Evidence for the role of OXT in regulating anxiety is undeniable. We expect that the diverse particularities of the OXT system will help broaden our understanding of anxiety and stress-related disorders. We conclude that OXT promises an enticing treatment option for human anxiety disorders especially those associated with socio-emotional dysfunctions.
Collapse
Affiliation(s)
- Wadih Jean Naja
- Department of Psychiatry, Lebanese University, Beirut, Lebanon. .,Gharios Medical Center, Mount Lebanon Hospital, Hazmieh, Beirut, Lebanon.
| | - Michaelangelo Pietro Aoun
- Department of Psychiatry, Lebanese University, Beirut, Lebanon.,Gharios Medical Center, Mount Lebanon Hospital, Hazmieh, Beirut, Lebanon
| |
Collapse
|
44
|
Peters ST, Bowen MT, Bohrer K, McGregor IS, Neumann ID. Oxytocin inhibits ethanol consumption and ethanol-induced dopamine release in the nucleus accumbens. Addict Biol 2017; 22:702-711. [PMID: 26810371 DOI: 10.1111/adb.12362] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 12/04/2015] [Accepted: 12/07/2015] [Indexed: 11/29/2022]
Abstract
Alcohol (EtOH) is one of the most widely abused recreational drugs and is arguably the most harmful. However, current treatment options for alcohol-use disorders generally have limited efficacy and poor uptake in the community. In this context, the neuropeptide oxytocin (OXT) has emerged as a promising potential treatment option for a number of substance-use disorders, including alcoholism. The utility of OXT in reducing consumption of and craving for a wide range of substances may lie in its ability to modulate drug-induced neurochemical effects within the mesolimbic dopamine pathway. However, the impact of OXT on EtOH actions in this pathway has yet to be explored. Here, we reveal that an acute intracerebroventricular (icv) infusion of OXT (1 µg/5 µl) attenuated voluntary EtOH (20 percent) self-administration after chronic intermittent access to EtOH for 59 days (28 drinking sessions) in male Wistar rats. Next, we demonstrated that an acute intraperitoneal (ip) injection of EtOH (1.5 g/kg, 15 percent w/v) increased dopamine release within the nucleus accumbens in both EtOH-naive rats and rats that had received 10 daily ip injections of EtOH. Icv OXT completely blocked the EtOH-induced dopamine release in both EtOH-naive and chronically treated rats. The attenuation of EtOH-induced dopamine release by OXT may help to explain the reduced EtOH self-administration observed following icv OXT infusion.
Collapse
Affiliation(s)
- Sebastian T. Peters
- Department of Behavioral and Molecular Neurobiology; University of Regensburg; Germany
- Current address: Department of Neurology; University Clinic Regensburg; Germany
| | | | - Kathrin Bohrer
- Department of Behavioral and Molecular Neurobiology; University of Regensburg; Germany
| | | | - Inga D. Neumann
- Department of Behavioral and Molecular Neurobiology; University of Regensburg; Germany
| |
Collapse
|
45
|
Molecular Basis of Oxytocin Receptor Signalling in the Brain: What We Know and What We Need to Know. Curr Top Behav Neurosci 2017; 35:3-29. [PMID: 28812263 DOI: 10.1007/7854_2017_6] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oxytocin (OT), a hypothalamic neuropeptide involved in regulating the social behaviour of all vertebrates, has been proposed as a treatment for a number of neuropsychiatric disorders characterised by deficits in the social domain. Over the last few decades, advances focused on understanding the social effects of OT and its role in physiological conditions and brain diseases, but much less has been done to clarify the molecular cascade of events involved in mediating such effects and in particular the cellular and molecular pharmacology of OT and its target receptor (OTR) in neuronal and glial cells.The entity and persistence of OT activity in the brain is closely related to the expression and regulation of the OTR expressed on the cell surface, which transmits the signal intracellularly and permits OT to affect cell function. Understanding the various signalling mechanisms mediating OTR-induced cell responses is crucial to determine the different responses in different cells and brain regions, and the success of OT and OT-derived analogues in the treatment of neurodevelopmental and psychiatric diseases depends on how well we can control such responses. In this review, we will consider the most important aspects of OT/OTR signalling by focusing on the molecular events involved in OT binding and coupling, on the main signalling pathways activated by the OTR in neuronal cells and on intracellular and plasma membrane OTR trafficking, all of which contribute to the quantitative and qualitative features of OT responses in the brain.
Collapse
|
46
|
Poisbeau P, Grinevich V, Charlet A. Oxytocin Signaling in Pain: Cellular, Circuit, System, and Behavioral Levels. Curr Top Behav Neurosci 2017; 35:193-211. [PMID: 28942595 DOI: 10.1007/7854_2017_14] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Originally confined to the initiation of parturition and milk ejection after birth, the hypothalamic nonapeptide oxytocin (OT) is now recognized as a critical determinant of social behavior and emotional processing. It accounts for the modulation of sensory processing and pain perception as OT displays a potent analgesic effect mediated by OT receptors (OTRs) expressed in the peripheral and central nervous systems. In our chapter, we will first systemically analyze known efferent and afferent OT neuron projections, which form the anatomical basis for OT modulation of somatosensory and pain processing. Next, we will focus on the synergy of distinct types of OT neurons (e.g., magno- and parvocellular OT neurons) which efficiently control acute inflammatory pain perception. Finally, we will describe how OT signaling mechanisms in the spinal cord control nociception, as well as how OT is able to modulate emotional pain processing within the central amygdala. In the conclusions at the end of the chapter, we will formulate perspectives in the study of OT effects on pain anticipation and pain memory, as well as propose some reasons for the application of exogenous OT for the treatment of certain types of pain in human patients.
Collapse
Affiliation(s)
- Pierrick Poisbeau
- Centre National de la Recherche Scientifique, University of Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France
| | - Valery Grinevich
- Schaller Research Group on Neuropeptides, German Cancer Research Center, University of Heidelberg, Heidelberg, BW, Germany.,Central Institute of Mental Health, University of Heidelberg, Mannheim, BW, Germany
| | - Alexandre Charlet
- Centre National de la Recherche Scientifique, University of Strasbourg, Institut des Neurosciences Cellulaires et Intégratives, Strasbourg, France. .,University of Strasbourg Institute for Advanced Study, USIAS, Strasbourg, France.
| |
Collapse
|
47
|
Chandaka GK, Wang L, Senogles S, Armstrong WE. Late Pregnancy is a Critical Period for Changes in Phosphorylated Mitogen-Activated Protein Kinase/Extracellular Signal-Regulated Kinase 1/2 in Oxytocin Neurones. J Neuroendocrinol 2016; 28:10.1111/jne.12398. [PMID: 27203238 PMCID: PMC5028259 DOI: 10.1111/jne.12398] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 04/28/2016] [Accepted: 05/16/2016] [Indexed: 12/19/2022]
Abstract
The physiological demands of parturition and lactation lead to the increased pulsatile release of oxytocin (OT) into the circulation from the neurohypophysial axons of OT neurones in the supraoptic (SON) and paraventricular (PVN) nuclei. These states of increased OT release are accompanied by a significant plasticity in magnocellular OT neurones and their synaptic connections, and many of these changes require activation of a central OT receptor. The mitogen-activated protein kinase/extracellular signal-regulated kinase pathway (MAPK/ERK) is assumed to be up-regulated in the PVN during lactation, and many of the effects of OT in peripheral and brain tissue are mediated through a MAPK/ERK pathway. The present study investigated whether this pathway is altered in the SON and PVN during late pregnancy [embryonic day (E)20-21], which is a critical period for OT plasticity induction, and for lactation, when plastic changes are sustained. Based on immunoreactivity for phosphorylated ERK1/2 (pERK1/2), the results suggest an enhanced activation of MAPK/ERK pathway in OT neurones specifically during late pregnancy in both the SON and PVN. Although immunoblots from the SON confirm this pregnancy-associated up-regulation in late pregnancy, they also suggest enhancement into lactation as well. Together, the results suggest an important role for the MAPK/ERK pathway during reproductive changes in the SON and PVN.
Collapse
Affiliation(s)
- G. K. Chandaka
- Department of Anatomy and NeurobiologyUniversity of Tennessee Health Science CenterMemphisTNUSA
| | - L. Wang
- Department of Anatomy and NeurobiologyUniversity of Tennessee Health Science CenterMemphisTNUSA
| | - S. Senogles
- Microbiology, Immunology and BiochemistryUniversity of Tennessee Health Science CenterMemphisTNUSA
- Neuroscience InstituteUniversity of Tennessee Health Science CenterMemphisTNUSA
| | - W. E. Armstrong
- Department of Anatomy and NeurobiologyUniversity of Tennessee Health Science CenterMemphisTNUSA
- Neuroscience InstituteUniversity of Tennessee Health Science CenterMemphisTNUSA
| |
Collapse
|
48
|
Abstract
Successfully rearing young places multiple demands on the mammalian female. These are met by a wide array of alterations in maternal physiology and behavior that are coordinated with the needs of the developing young, and include adaptations in neuroendocrine systems not directly involved in maternal behavior or lactation. In this article, attenuations in the behavioral and neuroendocrine responses to stressors, the alterations in metabolic pathways facilitating both increased food intake and conservation of energy, and the changes in fertility that occur postpartum are described. The mechanisms underlying these processes as well as the factors that contribute to them and the relative contributions of these stimuli at different times postpartum are also reviewed. The induction and maintenance of the adaptations observed in the postpartum maternal brain are dependent on mother-young interaction and, in most cases, on suckling stimulation and its consequences for the hormonal profile of the mother. The peptide hormone prolactin acting on receptors within the brain makes a major contribution to changes in metabolic pathways, suppression of fertility and the attenuation of the neuroendocrine response to stress during lactation. Oxytocin is also released, both into the circulation and in some hypothalamic nuclei, in response to suckling stimulation and this hormone has been implicated in the decrease in anxiety behavior seen in the early postpartum period. The relative importance of these hormones changes across lactation and it is becoming increasingly clear that many of the adaptations to motherhood reviewed here reflect the outcome of multiple influences. © 2016 American Physiological Society. Compr Physiol 6:1493-1518, 2016.
Collapse
Affiliation(s)
- Barbara Woodside
- Center for Studies in Behavioral Neurobiology, Department of Psychology, Concordia University, Montreal, Quebec, Canada
| |
Collapse
|
49
|
Abstract
The neuropeptide oxytocin (OXT) has been revealed as a profound anxiolytic and antistress factor of the brain, besides its many prosocial and reproductive effects. Therefore, there is substantial scientific and medical interest in its potential therapeutic use for the treatment of psychopathologies associated with anxiety, fear, and social dysfunctions, such as generalized anxiety disorder, posttraumatic stress disorder, and social anxiety disorder, as well as autism and schizophrenia, among others. Focusing on preclinical studies, we review the existing evidence for the regulatory capacity of OXT to fine-tune general and social anxiety-related behaviors, as well as cued and social fear conditioning from a translational perspective. The available evidence from animal and human studies substantiates the hypothesis of an imbalance of the endogenous brain OXT system in the etiology of anxiety disorders, particularly those with a social component such as social anxiety disorder. In addition, such an imbalance of the OXT system is also likely to be the consequence of chronic OXT treatment resulting in a dose-dependent reduction in OXT receptor availability and increased anxiety.
Collapse
|
50
|
Wang Y, Zhao S, Wu Z, Feng Y, Zhao C, Zhang C. Oxytocin in the regulation of social behaviours in medial amygdala-lesioned mice via the inhibition of the extracellular signal-regulated kinase signalling pathway. Clin Exp Pharmacol Physiol 2016; 42:465-74. [PMID: 25707920 DOI: 10.1111/1440-1681.12378] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 01/27/2015] [Accepted: 02/06/2015] [Indexed: 01/22/2023]
Abstract
The neuropeptide oxytocin (OXT) has been implicated in the pathophysiology of behavioural deficits among patients with autism spectrum disorder (ASD). However, the molecular mechanisms underlying its role in ASD remain unclear. In the present study, a murine model with ASD-like phenotypes was induced by intra-medial amygdala injection of N-methyl-d-aspartate, and it was used to investigate the role of OXT in behaviour regulation. Behavioural tests were performed to verify the ASD-like phenotypes of N-methyl-d-aspartate-treated mice, and the results showed that mice with bilateral medial amygdala lesions presented significant behavioural deficits, including impaired learning and memory and increased anxiety and depression. We also observed a notably decreased level of OXT in both the plasma and the hypothalamus of medial amygdala-lesioned mice, and the extracellular signal-regulated kinase (ERK) was activated. Further studies demonstrated that the administration of OXT alleviated ASD-like symptoms and significantly inhibited phosphorylation of ERK; the inhibitory effect was similar to that of U0126, an ERK signalling inhibitor. In addition, OXT administration modulated the expression of downstream proteins of the ERK signalling pathway, such as cyclic adenosine monophosphate response element binding and c-fos. Taken together, our data indicate that OXT plays an important role in ameliorating behavioural deficits in an ASD-like mouse model, which was mediated by inhibiting the ERK signalling pathway and its downstream proteins.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | | | | | | | | | | |
Collapse
|