1
|
El-Kadi RA, Sedeek MS, Abdelkader NF, Zaki HF, Kamel AS. Ameliorative Effect of Moringa oleifera Against CUMS-Induced Anxiety in Rats: β-Catenin and 5-HT 1 A Crosstalk. Mol Neurobiol 2025:10.1007/s12035-025-04911-8. [PMID: 40266546 DOI: 10.1007/s12035-025-04911-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/01/2025] [Indexed: 04/24/2025]
Abstract
Serotonin 1 A receptor (5-HT1 AR) signaling is pivotal for stress response, determining vulnerability or resilience to psychopathology. However, the precise pathological mechanisms underlying its role remain inconsistent. Moringa oleifera (MO), a plant with purported medicinal properties, has demonstrated potential efficacy against psychiatric disorders. However, no available information exists regarding its effects on 5-HT1 A signaling under normal and stressed conditions. This study is aimed at elucidating the effects of MO in conjunction with 5-HT1 A signaling. Rats were randomly assigned to four groups: normal (NRML), normal rats receiving MO orally at 200 mg/kg (MO), rats exposed to chronic unpredictable mild stress (CUMS) for 21 days (CUMS), and stressed rats administered MO from day 15 (CUMS + MO). Behavioral analysis was conducted using forced swimming and open field tests. Serotonergic markers, β-catenin, p-Erk, c-myc, and mTOR were assessed via ELISA, while miRNA clusters and individual miRNAs were analyzed using PCR. No significant differences were observed between the NRML and MO groups, both of which exhibited approximately normal biochemical activity, except for a decreased 5-HIAA/5-HT ratio in the MO group, which was reflected behaviorally. Rats subjected to CUMS displayed defective β-catenin signaling, potentially leading to compensatory activation of 5-HT1 A. Consistently, the CUMS + MO group exhibited normalized 5-HT1 A and 5-HT signaling, accompanied by reduced pThr183-Erk and its downstream targets, c-myc and miR- 203, to mitigate pathological anxiety. Additionally, mTOR and its downstream target, miR- 217, were reduced compared to stressed rats. MO exhibited a promising anxiolytic effect by modulating 5-HT1 A signaling, as evidenced by improved neurobehavioral outcomes and restoring biochemical balance in stressed rats. These findings highlight its potential therapeutic role in anxiety management.
Collapse
Affiliation(s)
- Rana A El-Kadi
- Alexandria University Hospitals, Champollion Street, El-Khartoum Square, El Azareeta, Alexandria City, 21131, Egypt
| | - Mohamed S Sedeek
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini, Cairo City, 11562, Egypt
- Pharmacognosy Department, Faculty of Pharmacy, King Salman International University, Ras-Sedr, South Sinai City, 46612, Egypt
| | - Noha F Abdelkader
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini, Cairo City, 11562, Egypt.
| | - Hala F Zaki
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini, Cairo City, 11562, Egypt
| | - Ahmed S Kamel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini, Cairo City, 11562, Egypt
| |
Collapse
|
2
|
Buenrostro-Jáuregui MH, Muñóz-Sánchez S, Rojas-Hernández J, Alonso-Orozco AI, Vega-Flores G, Tapia-de-Jesús A, Leal-Galicia P. A Comprehensive Overview of Stress, Resilience, and Neuroplasticity Mechanisms. Int J Mol Sci 2025; 26:3028. [PMID: 40243691 PMCID: PMC11988468 DOI: 10.3390/ijms26073028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Stress is a core concept in the mental health field that expands upon the seminal definition of stress as an acute response to the disruption of homeostasis. Stress is a complex process that involves both environmental challenges and the triggering of internal responses and impacts physiological, psychological, and behavioral systems. The capacity of the human brain to cope with stress is particularly crucial in early life, when neurodevelopment is highly plastic. Early-life stress (ELS), defined as exposure to severe chronic stress during sensitive periods of development, has been shown to cause lasting changes in brain structure and function. However, not all individuals exposed to ELS develop pathological outcomes, suggesting the presence of resilience mechanisms: adaptive processes that allow an individual to cope with adverse situations while maintaining psychological and neurobiological health. The aim of this review was to synthesize recent advances in the understanding of the neuroplasticity mechanisms underlying resilience to ELS. We discussed the neurobiological pathways implicated in stress response and adaptation, including the roles of neurogenesis, synaptic plasticity, and neural circuit remodeling. By focusing on the interplay between stress-induced neuroplastic changes and resilience mechanisms, we aimed to provide insights into potential therapeutic targets for stress-related psychopathology.
Collapse
Affiliation(s)
- Mario Humberto Buenrostro-Jáuregui
- Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (S.M.-S.); (J.R.-H.); (A.I.A.-O.); (P.L.-G.)
| | - Sinuhé Muñóz-Sánchez
- Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (S.M.-S.); (J.R.-H.); (A.I.A.-O.); (P.L.-G.)
| | - Jorge Rojas-Hernández
- Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (S.M.-S.); (J.R.-H.); (A.I.A.-O.); (P.L.-G.)
| | - Adriana Ixel Alonso-Orozco
- Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (S.M.-S.); (J.R.-H.); (A.I.A.-O.); (P.L.-G.)
- Facultad de Psicología, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - German Vega-Flores
- Ciencias de la Salud, Universidad Internacional de Valencia, 46002 Valencia, Spain;
- Educación, Universidad Internacional de La Rioja, 26006 Logroño, Spain
| | - Alejandro Tapia-de-Jesús
- Departamento de Salud, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico;
| | - Perla Leal-Galicia
- Departamento de Psicología, Universidad Iberoamericana Ciudad de México, Ciudad de México 01219, Mexico; (S.M.-S.); (J.R.-H.); (A.I.A.-O.); (P.L.-G.)
| |
Collapse
|
3
|
Holsboer F, Ising M. Precision Psychiatry Approach to Treat Depression and Anxiety Targeting the Stress Hormone System - V1b-antagonists as a Case in Point. PHARMACOPSYCHIATRY 2024; 57:263-274. [PMID: 39159843 DOI: 10.1055/a-2372-3549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
The future of depression pharmacotherapy lies in a precision medicine approach that recognizes that depression is a disease where different causalities drive symptoms. That approach calls for a departure from current diagnostic categories, which are broad enough to allow adherence to the "one-size-fits-all" paradigm, which is complementary to the routine use of "broad-spectrum" mono-amine antidepressants. Similar to oncology, narrowing the overinclusive diagnostic window by implementing laboratory tests, which guide specifically targeted treatments, will be a major step forward in overcoming the present drug discovery crisis.A substantial subgroup of patients presents with signs and symptoms of hypothalamic-pituitary-adrenocortical (HPA) overactivity. Therefore, this stress hormone system was considered to offer worthwhile targets. Some promising results emerged, but in sum, the results achieved by targeting corticosteroid receptors were mixed.More specific are non-peptidergic drugs that block stress-responsive neuropeptides, corticotropin-releasing hormone (CRH), and arginine vasopressin (AVP) in the brain by antagonizing their cognate CRHR1-and V1b-receptors. If a patient's depressive symptomatology is driven by overactive V1b-signaling then a V1b-receptor antagonist should be first-line treatment. To identify the patient having this V1b-receptor overactivity, a neuroendocrine test, the so-called dex/CRH-test, was developed, which indicates central AVP release but is too complicated to be routinely used. Therefore, this test was transformed into a gene-based "near-patient" test that allows immediate identification if a depressed patient's symptomatology is driven by overactive V1b-receptor signaling. We believe that this precision medicine approach will be the next major innovation in the pharmacotherapy of depression.
Collapse
Affiliation(s)
- Florian Holsboer
- Max Planck Institute of Psychiatry, Munich, Germany
- HMNC Holding GmbH, Munich, Germany
| | - Marcus Ising
- Max Planck Institute of Psychiatry, Munich, Germany
| |
Collapse
|
4
|
Arakawa H, Tokashiki M, Higuchi Y, Konno T. Adolescent social isolation disrupts developmental tuning of neuropeptide circuits in the hypothalamus to amygdala regulating social and defensive behavior. Peptides 2024; 175:171178. [PMID: 38368908 DOI: 10.1016/j.peptides.2024.171178] [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: 01/04/2024] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 02/20/2024]
Abstract
Engaging in positive social (i.e., prosocial) interactions during adolescence acts to modulate neural circuits that determine adult adaptive behavior. While accumulating evidence indicates that a strong craving for prosocial behavior contributes to sustaining neural development, the consequences of social deprivation during adolescence on social neural circuits, including those involving oxytocin (OXT) and vasopressin (AVP), are poorly characterized. We evaluated adaptive behaviors in socially isolated mice, including anxiety-like, social, and defensive behaviors, along with OXT and AVP neural profiles in relevant brain regions. Social isolation from postnatal day (P-)22 to P-48 induced enhanced defensive and exploratory behaviors, in nonsocial and social contexts. Unlike OXT neurons, AVP+ cell density in the paraventricular nucleus of the hypothalamus increases with age in males. Social isolation also modulated gene expression in the medial amygdala (MeA), including the upregulation of OXT receptors in males and the downregulation of AVP1a receptors in both sexes. Socially isolated mice showed an enhanced defensive, anogenital approach toward a novel adult female during direct social interactions. Subsequent c-Fos mapping revealed diminished neural activity in restricted brain areas, including the MeA, lateral septum, and posterior intralaminar nucleus of the thalamus, in socially isolated mice. These data indicate that neural signals arising from daily social interactions invoke region-specific modification of neuropeptide expression that coordinates with altered defensiveness and neural responsivities, including OXT- and AVP-projecting regions. The present findings indicate an involvement of OXT and AVP circuits in adolescent neural and behavioral plasticity that is tuned by daily social interaction.
Collapse
Affiliation(s)
- Hiroyuki Arakawa
- Department of Pharmacology, University of Michigan School of Medicine, MI, USA.
| | - Mana Tokashiki
- Faculty of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Yuki Higuchi
- Department of Systems Physiology, University of the Ryukyus Graduate School of Medicine, Okinawa, Japan
| | - Toshihiro Konno
- Department of Subtropical Agro-Environmental Sciences, Faculty of Agriculture, University of the Ryukyus, Okinawa, Japan; The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
5
|
Rigney N, de Vries GJ, Petrulis A. Modulation of social behavior by distinct vasopressin sources. Front Endocrinol (Lausanne) 2023; 14:1127792. [PMID: 36860367 PMCID: PMC9968743 DOI: 10.3389/fendo.2023.1127792] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 01/30/2023] [Indexed: 02/15/2023] Open
Abstract
The neuropeptide arginine-vasopressin (AVP) is well known for its peripheral effects on blood pressure and antidiuresis. However, AVP also modulates various social and anxiety-related behaviors by its actions in the brain, often sex-specifically, with effects typically being stronger in males than in females. AVP in the nervous system originates from several distinct sources which are, in turn, regulated by different inputs and regulatory factors. Based on both direct and indirect evidence, we can begin to define the specific role of AVP cell populations in social behavior, such as, social recognition, affiliation, pair bonding, parental behavior, mate competition, aggression, and social stress. Sex differences in function may be apparent in both sexually-dimorphic structures as well as ones without prominent structural differences within the hypothalamus. The understanding of how AVP systems are organized and function may ultimately lead to better therapeutic interventions for psychiatric disorders characterized by social deficits.
Collapse
Affiliation(s)
- Nicole Rigney
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | | | | |
Collapse
|
6
|
Salivary Antioxidant Capacity and Magnesium in Generalized Anxiety Disorder. Metabolites 2023; 13:metabo13010073. [PMID: 36676998 PMCID: PMC9862115 DOI: 10.3390/metabo13010073] [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: 11/14/2022] [Revised: 12/29/2022] [Accepted: 12/29/2022] [Indexed: 01/22/2023] Open
Abstract
Generalized anxiety disorder (GAD) is a prevalent disorder. The search for biomarkers may contribute to new knowledge about molecular pathogenesis and treatment. Since oxidative stress and micronutrient imbalance play a key role in the development of mental disorders, we aimed to study salivary antioxidant capacity and magnesium in patients with GAD in an anxiety model of solving problems with increasing complexity. The study subgroup consisted of 15 patients with GAD, and 17 healthy volunteers of the same age made up the control subgroup. Participants took a test with six levels of difficulty, which included false feedback. In this test, the participants were asked to remember the colors of balloons and react when the color changed. The reaction time, the number of correct answers, as well as biochemical parameters such as the antioxidant capacity of saliva and salivary magnesium, were assessed. There was no difference in the results of the quest between the subgroups; however, anxious participants spent more time at the moment of experimental frustration due to incorrect feedback and additional negative psycho-emotional load. Antioxidant capacity did not differ between the subgroups both before and after the experimental session. Average antioxidant capacity also did not change significantly at the endpoint of the experiment. However, the endpoint antioxidant capacity correlated negatively with the reaction time in anxious patients in the second block (where the false feedback as a frustrating factor appeared). Magnesium was initially significantly higher in the group of anxious participants and decreased at the experiment endpoint; in healthy patients, there were no changes in salivary magnesium at the endpoint. In conclusion, the compensatory potential of oxidative metabolism and magnesium in patients with GAD was spent with additional psycho-emotional stress, in contrast to healthy individuals, but it was sufficient to avoid exhaustion during experimental frustrating exposure.
Collapse
|
7
|
Arakawa H, Higuchi Y. Exocrine scent marking: Coordinative role of arginine vasopressin in the systemic regulation of social signaling behaviors. Neurosci Biobehav Rev 2022; 136:104597. [PMID: 35248677 DOI: 10.1016/j.neubiorev.2022.104597] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 03/01/2022] [Accepted: 03/01/2022] [Indexed: 12/25/2022]
Abstract
Arginine vasopressin (AVP) is a neurohypophysial hormone that coordinatively regulates central socio-emotional behavior and peripheral control of antidiuretic fluid homeostasis. Most mammals, including rodents, utilize exocrine or urine-contained scent marking as a social signaling tool that facilitates social adaptation. The exocrine scent marking behavior is postulated to fine-tune sensory and cognitive abilities to recognize key social features via exocrine/urinary olfactory cues and subsequently control exocrine deposition or urinary marking through the mediation of osmotic fluid balance. AVP is implicated as a major player in controlling both recognition and signaling responses. This review provides constructive hypotheses on the coordinative processes of the AVP neurohypophysial circuits in the systemic regulations of fluid control and social-communicative behavior, via the expression of exocrine scent marking, and further emphasizes a potential role of AVP in a common mechanism underlying social communication in rodents.
Collapse
Affiliation(s)
- Hiroyuki Arakawa
- Depertment of Systems Physiology, University of the Ryukyus School of Medicine, Okinawa, Japan.
| | - Yuki Higuchi
- Depertment of Systems Physiology, University of the Ryukyus School of Medicine, Okinawa, Japan
| |
Collapse
|
8
|
Rigney N, Whylings J, de Vries GJ, Petrulis A. Sex Differences in the Control of Social Investigation and Anxiety by Vasopressin Cells of the Paraventricular Nucleus of the Hypothalamus. Neuroendocrinology 2021; 111:521-535. [PMID: 32541145 PMCID: PMC7736187 DOI: 10.1159/000509421] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022]
Abstract
The neuropeptide arginine-vasopressin (AVP) has long been implicated in the regulation of social behavior and communication in diverse taxa, but the source of AVP release relevant for behavior has not been precisely determined. Potential sources include hypothalamic cell populations such as the paraventricular (PVN), supraoptic, and suprachiasmatic nuclei, as well as extrahypothalamic cell groups in the extended amygdala. To address if AVP-expressing cells in the PVN are important for mouse social communication, we deleted PVN AVP-expressing cells using viral-mediated delivery of Cre-dependent caspase-9 cell death construct into the PVN of AVP-Cre-positive mice (expressing Cre-recombinase under the control of the AVP promoter) or AVP-Cre-negative littermate controls, and assessed their levels of social investigation, social communication, anxiety, sex behavior, and aggressive behavior. We found that these lesions increased social investigation in females, but not in males. However, in males but not in females, these lesions increased non-social anxiety-related behaviors in the elevated-plus maze. These results therefore point at differential involvement of PVN AVP-expressing cells in the context of social and emotional behavior in the two sexes, which may contribute to sex differences in social communication and anxiety disorders.
Collapse
Affiliation(s)
- Nicole Rigney
- Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA,
| | - Jack Whylings
- Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Geert J de Vries
- Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Aras Petrulis
- Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| |
Collapse
|
9
|
Whylings J, Rigney N, de Vries GJ, Petrulis A. Removal of vasopressin cells from the paraventricular nucleus of the hypothalamus enhances lipopolysaccharide-induced sickness behaviour in mice. J Neuroendocrinol 2021; 33:e12915. [PMID: 33617060 PMCID: PMC8543850 DOI: 10.1111/jne.12915] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/07/2020] [Accepted: 10/20/2020] [Indexed: 12/26/2022]
Abstract
Vasopressin (AVP) cells in the paraventricular nucleus of the hypothalamus (PVN) are activated during sickness and project to multiple nuclei responsible for the anxiety, social and motivated behaviours affected during sickness, suggesting that these cells may play a role in sickness behaviours, typically expressed as reduced mobility, increased anxiety, anhedonia and social withdrawal. In the present study, we selectively ablated AVP neurones in the PVN of male and female mice (Mus musculus) and induced sickness behaviour via injection of bacterial lipopolysaccharide (LPS). We found that PVN AVP ablation increased the effects of LPS, specifically by further decreasing sucrose preference in males and females and decreasing the social preference of males, monitored within 24 hours of LPS injection. These results suggest that PVN AVP contributes to the change in motivated behaviours during sickness and may help promote recovery from infection..
Collapse
Affiliation(s)
- Jack Whylings
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Nicole Rigney
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| | - Geert J de Vries
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - Aras Petrulis
- Neuroscience Institute, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
10
|
Fedorov VN, Koroleva SV, Zubova TA, Andreeva LA, Myasoedov NF. Preparations Based on Regulatory Peptides—a New Class of Medicines. NEUROCHEM J+ 2020. [DOI: 10.1134/s1819712420040121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
11
|
Carter CS, Kenkel WM, MacLean EL, Wilson SR, Perkeybile AM, Yee JR, Ferris CF, Nazarloo HP, Porges SW, Davis JM, Connelly JJ, Kingsbury MA. Is Oxytocin "Nature's Medicine"? Pharmacol Rev 2020; 72:829-861. [PMID: 32912963 PMCID: PMC7495339 DOI: 10.1124/pr.120.019398] [Citation(s) in RCA: 230] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Oxytocin is a pleiotropic, peptide hormone with broad implications for general health, adaptation, development, reproduction, and social behavior. Endogenous oxytocin and stimulation of the oxytocin receptor support patterns of growth, resilience, and healing. Oxytocin can function as a stress-coping molecule, an anti-inflammatory, and an antioxidant, with protective effects especially in the face of adversity or trauma. Oxytocin influences the autonomic nervous system and the immune system. These properties of oxytocin may help explain the benefits of positive social experiences and have drawn attention to this molecule as a possible therapeutic in a host of disorders. However, as detailed here, the unique chemical properties of oxytocin, including active disulfide bonds, and its capacity to shift chemical forms and bind to other molecules make this molecule difficult to work with and to measure. The effects of oxytocin also are context-dependent, sexually dimorphic, and altered by experience. In part, this is because many of the actions of oxytocin rely on its capacity to interact with the more ancient peptide molecule, vasopressin, and the vasopressin receptors. In addition, oxytocin receptor(s) are epigenetically tuned by experience, especially in early life. Stimulation of G-protein-coupled receptors triggers subcellular cascades allowing these neuropeptides to have multiple functions. The adaptive properties of oxytocin make this ancient molecule of special importance to human evolution as well as modern medicine and health; these same characteristics also present challenges to the use of oxytocin-like molecules as drugs that are only now being recognized. SIGNIFICANCE STATEMENT: Oxytocin is an ancient molecule with a major role in mammalian behavior and health. Although oxytocin has the capacity to act as a "natural medicine" protecting against stress and illness, the unique characteristics of the oxytocin molecule and its receptors and its relationship to a related hormone, vasopressin, have created challenges for its use as a therapeutic drug.
Collapse
Affiliation(s)
- C Sue Carter
- Kinsey Institute, Indiana University, Bloomington, Indiana (C.S.C., W.M.K., A.M.P., H.P.N., S.W.P.); School of Anthropology, Department of Psychology, and College of Veterinary Medicine, University of Arizona, Tucson, Arizona (E.L.M.); Department of Chemistry, University of Oslo, Oslo, Norway (S.R.W.); Institute of Animal Welfare Science, University of Veterinary Medicine, Vienna, Austria (J.R.Y.); Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (C.F.F.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (J.M.D.); Department of Psychology, University of Virginia, Charlottesville, Virginia (J.J.C.); and Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charleston, Massachusetts (M.A.K.)
| | - William M Kenkel
- Kinsey Institute, Indiana University, Bloomington, Indiana (C.S.C., W.M.K., A.M.P., H.P.N., S.W.P.); School of Anthropology, Department of Psychology, and College of Veterinary Medicine, University of Arizona, Tucson, Arizona (E.L.M.); Department of Chemistry, University of Oslo, Oslo, Norway (S.R.W.); Institute of Animal Welfare Science, University of Veterinary Medicine, Vienna, Austria (J.R.Y.); Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (C.F.F.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (J.M.D.); Department of Psychology, University of Virginia, Charlottesville, Virginia (J.J.C.); and Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charleston, Massachusetts (M.A.K.)
| | - Evan L MacLean
- Kinsey Institute, Indiana University, Bloomington, Indiana (C.S.C., W.M.K., A.M.P., H.P.N., S.W.P.); School of Anthropology, Department of Psychology, and College of Veterinary Medicine, University of Arizona, Tucson, Arizona (E.L.M.); Department of Chemistry, University of Oslo, Oslo, Norway (S.R.W.); Institute of Animal Welfare Science, University of Veterinary Medicine, Vienna, Austria (J.R.Y.); Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (C.F.F.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (J.M.D.); Department of Psychology, University of Virginia, Charlottesville, Virginia (J.J.C.); and Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charleston, Massachusetts (M.A.K.)
| | - Steven R Wilson
- Kinsey Institute, Indiana University, Bloomington, Indiana (C.S.C., W.M.K., A.M.P., H.P.N., S.W.P.); School of Anthropology, Department of Psychology, and College of Veterinary Medicine, University of Arizona, Tucson, Arizona (E.L.M.); Department of Chemistry, University of Oslo, Oslo, Norway (S.R.W.); Institute of Animal Welfare Science, University of Veterinary Medicine, Vienna, Austria (J.R.Y.); Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (C.F.F.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (J.M.D.); Department of Psychology, University of Virginia, Charlottesville, Virginia (J.J.C.); and Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charleston, Massachusetts (M.A.K.)
| | - Allison M Perkeybile
- Kinsey Institute, Indiana University, Bloomington, Indiana (C.S.C., W.M.K., A.M.P., H.P.N., S.W.P.); School of Anthropology, Department of Psychology, and College of Veterinary Medicine, University of Arizona, Tucson, Arizona (E.L.M.); Department of Chemistry, University of Oslo, Oslo, Norway (S.R.W.); Institute of Animal Welfare Science, University of Veterinary Medicine, Vienna, Austria (J.R.Y.); Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (C.F.F.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (J.M.D.); Department of Psychology, University of Virginia, Charlottesville, Virginia (J.J.C.); and Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charleston, Massachusetts (M.A.K.)
| | - Jason R Yee
- Kinsey Institute, Indiana University, Bloomington, Indiana (C.S.C., W.M.K., A.M.P., H.P.N., S.W.P.); School of Anthropology, Department of Psychology, and College of Veterinary Medicine, University of Arizona, Tucson, Arizona (E.L.M.); Department of Chemistry, University of Oslo, Oslo, Norway (S.R.W.); Institute of Animal Welfare Science, University of Veterinary Medicine, Vienna, Austria (J.R.Y.); Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (C.F.F.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (J.M.D.); Department of Psychology, University of Virginia, Charlottesville, Virginia (J.J.C.); and Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charleston, Massachusetts (M.A.K.)
| | - Craig F Ferris
- Kinsey Institute, Indiana University, Bloomington, Indiana (C.S.C., W.M.K., A.M.P., H.P.N., S.W.P.); School of Anthropology, Department of Psychology, and College of Veterinary Medicine, University of Arizona, Tucson, Arizona (E.L.M.); Department of Chemistry, University of Oslo, Oslo, Norway (S.R.W.); Institute of Animal Welfare Science, University of Veterinary Medicine, Vienna, Austria (J.R.Y.); Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (C.F.F.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (J.M.D.); Department of Psychology, University of Virginia, Charlottesville, Virginia (J.J.C.); and Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charleston, Massachusetts (M.A.K.)
| | - Hossein P Nazarloo
- Kinsey Institute, Indiana University, Bloomington, Indiana (C.S.C., W.M.K., A.M.P., H.P.N., S.W.P.); School of Anthropology, Department of Psychology, and College of Veterinary Medicine, University of Arizona, Tucson, Arizona (E.L.M.); Department of Chemistry, University of Oslo, Oslo, Norway (S.R.W.); Institute of Animal Welfare Science, University of Veterinary Medicine, Vienna, Austria (J.R.Y.); Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (C.F.F.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (J.M.D.); Department of Psychology, University of Virginia, Charlottesville, Virginia (J.J.C.); and Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charleston, Massachusetts (M.A.K.)
| | - Stephen W Porges
- Kinsey Institute, Indiana University, Bloomington, Indiana (C.S.C., W.M.K., A.M.P., H.P.N., S.W.P.); School of Anthropology, Department of Psychology, and College of Veterinary Medicine, University of Arizona, Tucson, Arizona (E.L.M.); Department of Chemistry, University of Oslo, Oslo, Norway (S.R.W.); Institute of Animal Welfare Science, University of Veterinary Medicine, Vienna, Austria (J.R.Y.); Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (C.F.F.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (J.M.D.); Department of Psychology, University of Virginia, Charlottesville, Virginia (J.J.C.); and Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charleston, Massachusetts (M.A.K.)
| | - John M Davis
- Kinsey Institute, Indiana University, Bloomington, Indiana (C.S.C., W.M.K., A.M.P., H.P.N., S.W.P.); School of Anthropology, Department of Psychology, and College of Veterinary Medicine, University of Arizona, Tucson, Arizona (E.L.M.); Department of Chemistry, University of Oslo, Oslo, Norway (S.R.W.); Institute of Animal Welfare Science, University of Veterinary Medicine, Vienna, Austria (J.R.Y.); Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (C.F.F.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (J.M.D.); Department of Psychology, University of Virginia, Charlottesville, Virginia (J.J.C.); and Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charleston, Massachusetts (M.A.K.)
| | - Jessica J Connelly
- Kinsey Institute, Indiana University, Bloomington, Indiana (C.S.C., W.M.K., A.M.P., H.P.N., S.W.P.); School of Anthropology, Department of Psychology, and College of Veterinary Medicine, University of Arizona, Tucson, Arizona (E.L.M.); Department of Chemistry, University of Oslo, Oslo, Norway (S.R.W.); Institute of Animal Welfare Science, University of Veterinary Medicine, Vienna, Austria (J.R.Y.); Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (C.F.F.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (J.M.D.); Department of Psychology, University of Virginia, Charlottesville, Virginia (J.J.C.); and Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charleston, Massachusetts (M.A.K.)
| | - Marcy A Kingsbury
- Kinsey Institute, Indiana University, Bloomington, Indiana (C.S.C., W.M.K., A.M.P., H.P.N., S.W.P.); School of Anthropology, Department of Psychology, and College of Veterinary Medicine, University of Arizona, Tucson, Arizona (E.L.M.); Department of Chemistry, University of Oslo, Oslo, Norway (S.R.W.); Institute of Animal Welfare Science, University of Veterinary Medicine, Vienna, Austria (J.R.Y.); Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts (C.F.F.); Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois (J.M.D.); Department of Psychology, University of Virginia, Charlottesville, Virginia (J.J.C.); and Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Charleston, Massachusetts (M.A.K.)
| |
Collapse
|
12
|
Two ancient neuropeptides, PACAP and AVP, modulate motivated behavior at synapses in the extrahypothalamic brain: a study in contrast. Cell Tissue Res 2018; 375:103-122. [DOI: 10.1007/s00441-018-2958-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 11/05/2018] [Indexed: 01/24/2023]
|
13
|
Harper KM, Knapp DJ, Criswell HE, Breese GR. Vasopressin and alcohol: a multifaceted relationship. Psychopharmacology (Berl) 2018; 235:3363-3379. [PMID: 30392132 PMCID: PMC6286152 DOI: 10.1007/s00213-018-5099-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 10/28/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Arginine vasopressin (VP) has been implicated in a number of neuropsychiatric disorders with an emphasis on situations where stress increased the severity of the disorder. Based on this hypothesized role for VP in neuropsychiatric disorders, much research is currently being undertaken in humans and animals to test VP as a target for treatment of a number of these disorders including alcohol abuse. OBJECTIVES To provide a summary of the literature regarding the role of VP in alcohol- and stress-related behaviors including the use of drugs that target VP in clinical trials. RESULTS Changes in various components of the VP system occur with alcohol and stress. Manipulating VP or its receptors can alter alcohol- and stress-related behaviors including tolerance to alcohol, alcohol drinking, and anxiety-like behavior. Finally, the hypothalamic-pituitary-adrenal axis response to alcohol is also altered by manipulating the VP system. However, clinical trials of VP antagonists have had mixed results. CONCLUSIONS A review of VP's involvement in alcohol's actions demonstrates that there is much to be learned about brain regions involved in VP-mediated effects on behavior. Thus, future work should focus on elucidating relevant brain regions. By using previous knowledge of the actions of VP and determining the brain regions and/or systems involved in its different behavioral effects, it may be possible to identify a specific receptor subtype target, drug treatment combination, or specific clinical contexts that may point toward a more successful treatment.
Collapse
Affiliation(s)
- Kathryn M Harper
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB #7178, Thurston Bowles Building, Chapel Hill, NC, 27599-7178, USA.
| | - Darin J Knapp
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB #7178, Thurston Bowles Building, Chapel Hill, NC, 27599-7178, USA
- Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7178, USA
| | - Hugh E Criswell
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB #7178, Thurston Bowles Building, Chapel Hill, NC, 27599-7178, USA
- Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7178, USA
| | - George R Breese
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB #7178, Thurston Bowles Building, Chapel Hill, NC, 27599-7178, USA
- Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7178, USA
- Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599-7178, USA
| |
Collapse
|
14
|
Bayerl DS, Bosch OJ. Brain vasopressin signaling modulates aspects of maternal behavior in lactating rats. GENES BRAIN AND BEHAVIOR 2018; 18:e12517. [DOI: 10.1111/gbb.12517] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/22/2018] [Accepted: 09/11/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Doris S. Bayerl
- Department of Behavioural and Molecular Neurobiology; Regensburg Center of Neuroscience, University of Regensburg; Regensburg Germany
| | - Oliver J. Bosch
- Department of Behavioural and Molecular Neurobiology; Regensburg Center of Neuroscience, University of Regensburg; Regensburg Germany
| |
Collapse
|
15
|
Naik RR, Sotnikov SV, Diepold RP, Iurato S, Markt PO, Bultmann A, Brehm N, Mattheus T, Lutz B, Erhardt A, Binder EB, Schmidt U, Holsboer F, Landgraf R, Czibere L. Polymorphism in Tmem132d regulates expression and anxiety-related behavior through binding of RNA polymerase II complex. Transl Psychiatry 2018; 8:1. [PMID: 29317594 PMCID: PMC5802467 DOI: 10.1038/s41398-017-0025-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 07/30/2017] [Accepted: 08/29/2017] [Indexed: 01/16/2023] Open
Abstract
TMEM132D is a candidate gene, where risk genotypes have been associated with anxiety severity along with higher mRNA expression in the frontal cortex of panic disorder patients. Concurrently, in a high (HAB) and low (LAB) trait anxiety mouse model, Tmem132d was found to show increased expression in the anterior cingulate cortex (aCC) of HAB as compared to LAB mice. To understand the molecular underpinnings underlying the differential expression, we sequenced the gene and found two single-nucleotide polymorphisms (SNPs) in the promoter differing between both lines which could explain the observed mRNA expression profiles using gene reporter assays. In addition, there was no difference in basal DNA methylation in the CpG Island that encompasses the HAB vs. LAB Tmem132d promoter region. Furthermore, we found significantly higher binding of RNA polymerase II (POLR2A) to the proximal HAB-specific SNP (rs233264624) than the corresponding LAB locus in an oligonucleotide pull-down assay, suggesting increased transcription. Virus mediated overexpression of Tmem132d in the aCC of C57BL/6 J mice could confirm its role in mediating an anxiogenic phenotype. To model gene-environmental interactions, HAB mice exposed to enriched environment (HAB-EE) responded with decreased anxiety levels but, had enhanced Tmem132d mRNA expression as compared to standard-housed HAB (HAB-SH) mice. While LAB mice subjected to unpredictable chronic mild stress (LAB-UCMS) exhibited higher anxiety levels and had lower mRNA expression compared to standard-housed LAB (LAB-SH) mice. Chromatin immunoprecipitation revealed significantly higher binding of POLR2A to rs233264624 in HAB-EE, while LAB-UCMS had lower POLR2A binding at this locus, thus explaining the enhanced or attenuated expression of Tmem132d compared to their respective SH controls. To further investigate gene-environment interactions, DNA methylation was assessed using Illumina 450 K BeadChip in 74 panic disorder patients. Significant methylation differences were observed in two CpGs (cg26322591 and cg03283235) located in TMEM132D depending on the number of positive life events supporting the results of an influence of positive environmental cues on regulation of Tmem132d expression in mice.
Collapse
Affiliation(s)
- Roshan R Naik
- Max Planck Institute of Psychiatry, 80804, Munich, Germany.
- Department of Behavioral and Molecular Neurobiology, University of Regensburg, Regensburg, Germany.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 308232, Singapore.
| | - Sergey V Sotnikov
- Max Planck Institute of Psychiatry, 80804, Munich, Germany
- Department of Normal Physiology, Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Stella Iurato
- Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | | | | | - Nadine Brehm
- Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | - Tobias Mattheus
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | | | | | - Ulrike Schmidt
- Max Planck Institute of Psychiatry, 80804, Munich, Germany
| | | | | | - Ludwig Czibere
- Max Planck Institute of Psychiatry, 80804, Munich, Germany
- Labor Becker und, 81671, Munich, Germany
| |
Collapse
|
16
|
Wade M, Hoffmann TJ, Knafo-Noam A, O'Connor TG, Jenkins JM. Oxytocin and vasopressin hormone genes in children's externalizing problems: A cognitive endophenotype approach. Horm Behav 2016; 82:78-86. [PMID: 27155104 DOI: 10.1016/j.yhbeh.2016.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 03/04/2016] [Accepted: 05/02/2016] [Indexed: 10/21/2022]
Abstract
Externalizing problems are among the most common mental health problems of children. Research suggests that these problems are heritable, yet little is known about the specific genes involved in their pathophysiology. The current study examined a genotype-endophenotype-phenotype model of externalizing problems in 320 preschool-aged children. Markers of the oxytocin (OXT) and arginine vasopressin (AVP) hormone genes were selected as candidates owing to their known association with psychopathology in other domains. We tested whether OXT and AVP variants were related to children's externalizing problems, as well as two cognitive endophenotypes presumed to underlie these problems: theory of mind (ToM) and executive functioning (EF). Externalizing problems were assessed at age 4.5 using a previously-validated rating scale. ToM and EF were measured with age-appropriate tasks. Using a family-based association design and controlling for non-genomic confounds, support was found for an association between a two-marker OXT haplotype (rs2740210-rs2770378) and a two-marker AVP haplotype (rs1887854-rs3761249) and externalizing problems. Specific associations of these haplotypes with ToM and EF were also observed. Further, ToM and EF were shown to independently and jointly predict externalizing problems, and to partially mediate the effects of OXT and AVP on externalizing problems. This study provides the first evidence that genetic variation in OXT and AVP may contribute to individual differences in childhood externalizing problems, and that these effects may operate through emerging neurocognitive abilities in the preschool period.
Collapse
Affiliation(s)
- Mark Wade
- Department of Applied Psychology and Human Development, University of Toronto, Canada.
| | - Thomas J Hoffmann
- Department of Epidemiology and Biostatistics, and Institute for Human Genetics, University of California at San Francisco, USA
| | - Ariel Knafo-Noam
- Department of Psychology, The Hebrew University of Jerusalem, Israel
| | | | - Jennifer M Jenkins
- Department of Applied Psychology and Human Development, University of Toronto, Canada.
| |
Collapse
|
17
|
Nussbaumer M, Asara JM, Teplytska L, Murphy MP, Logan A, Turck CW, Filiou MD. Selective Mitochondrial Targeting Exerts Anxiolytic Effects In Vivo. Neuropsychopharmacology 2016; 41:1751-8. [PMID: 26567514 PMCID: PMC4869042 DOI: 10.1038/npp.2015.341] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 10/22/2015] [Accepted: 11/08/2015] [Indexed: 12/31/2022]
Abstract
Current treatment strategies for anxiety disorders are predominantly symptom-based. However, a third of anxiety patients remain unresponsive to anxiolytics highlighting the need for more effective, mechanism-based therapeutic approaches. We have previously compared high vs low anxiety mice and identified changes in mitochondrial pathways, including oxidative phosphorylation and oxidative stress. In this work, we show that selective pharmacological targeting of these mitochondrial pathways exerts anxiolytic effects in vivo. We treated high anxiety-related behavior (HAB) mice with MitoQ, an antioxidant that selectively targets mitochondria. MitoQ administration resulted in decreased anxiety-related behavior in HAB mice. This anxiolytic effect was specific for high anxiety as MitoQ treatment did not affect the anxiety phenotype of C57BL/6N and DBA/2J mouse strains. We furthermore investigated the molecular underpinnings of the MitoQ-driven anxiolytic effect and found that MitoQ treatment alters the brain metabolome and that the response to MitoQ treatment is characterized by distinct molecular signatures. These results indicate that a mechanism-driven approach based on selective mitochondrial targeting has the potential to attenuate the high anxiety phenotype in vivo, thus paving the way for translational implementation as long-term MitoQ administration is well-tolerated with no reported side effects in mice and humans.
Collapse
Affiliation(s)
| | - John M Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | - Michaela D Filiou
- Max Planck Institute of Psychiatry, Munich, Germany,Proteomics and Biomarkers, Max Planck Institute of Psychiatry, Kraepelinstr. 2, Munich 80804, Germany, Tel: +49 89 30622 506, Fax: +49 89 30622 200, E-mail:
| |
Collapse
|
18
|
Oxytocin Stimulates Extracellular Ca2+ Influx Through TRPV2 Channels in Hypothalamic Neurons to Exert Its Anxiolytic Effects. Neuropsychopharmacology 2015; 40:2938-47. [PMID: 26013963 PMCID: PMC4864629 DOI: 10.1038/npp.2015.147] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 05/02/2015] [Accepted: 05/04/2015] [Indexed: 11/09/2022]
Abstract
There is growing interest in anxiolytic and pro-social effects of the neuropeptide oxytocin (OXT), but the underlying intraneuronal mechanisms are largely unknown. Here we examined OXT-mediated anxiolysis in the hypothalamic paraventricular nucleus (PVN) of rats and effects of OXT administration on signaling events in hypothalamic primary and immortalized cells. In vivo, the application of SKF96365 prevented the anxiolytic activity of OXT in the PVN, suggesting that changes in intracellular Ca(2+) mediate the acute OXT behavioral effects. In vitro, mainly in the neurons with autonomous Ca(2+) oscillations, OXT increased intracellular Ca(2+) concentration and oscillation amplitude. Pharmacological intervention revealed OXT-dependent changes in Ca(2+) signaling that required activation of transient receptor potential vanilloid type-2 channel (TRPV2), mediated by phosphoinositide 3-kinase. TRPV2 induced the activation of the anxiolytic mitogen-activated protein kinase kinase (MEK1/2). In situ, immunohistochemistry revealed co-localization of TRPV2 and OXT in the PVN. Thus, functional and pharmacological analyses identified TRPV2 as a mediator of anxiolytic effects of OXT, conveying the OXT signal to MEK1/2 via modulation of intracellular Ca(2+).
Collapse
|
19
|
Selective breeding for high anxiety introduces a synonymous SNP that increases neuropeptide S receptor activity. J Neurosci 2015; 35:4599-613. [PMID: 25788677 DOI: 10.1523/jneurosci.4764-13.2015] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Neuropeptide S (NPS) has generated substantial interest due to its anxiolytic and fear-attenuating effects in rodents, while a corresponding receptor polymorphism associated with increased NPS receptor (NPSR1) surface expression and efficacy has been implicated in an increased risk of panic disorder in humans. To gain insight into this paradox, we examined the NPS system in rats and mice bred for high anxiety-related behavior (HAB) versus low anxiety-related behavior, and, thereafter, determined the effect of central NPS administration on anxiety- and fear-related behavior. The HAB phenotype was accompanied by lower basal NPS receptor (Npsr1) expression, which we could confirm via in vitro dual luciferase promoter assays. Assessment of shorter Npsr1 promoter constructs containing a sequence mutation that introduces a glucocorticoid receptor transcription factor binding site, confirmed via oligonucleotide pull-down assays, revealed increased HAB promoter activity-an effect that was prevented by dexamethasone. Analogous to the human NPSR1 risk isoform, functional analysis of a synonymous single nucleotide polymorphism in the coding region of HAB rodents revealed that it caused a higher cAMP response to NPS stimulation. Assessment of the behavioral consequence of these differences revealed that intracerebroventricular NPS reversed the hyperanxiety of HAB rodents as well as the impaired cued-fear extinction in HAB rats and the enhanced fear expression in HAB mice, respectively. These results suggest that alterations in the NPS system, conserved across rodents and humans, contribute to innate anxiety and fear, and that HAB rodents are particularly suited to resolve the apparent discrepancy between the preclinical and clinical findings to date.
Collapse
|
20
|
Abstract
The wide spectrum of disruptions that characterizes major depressive disorder (MDD) and bipolar disorder (BD) highlights the difficulties researchers are posed with as they try to mimic these disorders in the laboratory. Nonetheless, numerous attempts have been made to create rodent models of mood disorders or at least models of the symptoms of MDD and BD. Present antidepressants are all descendants of the serendipitous findings in the 1950s that the monoamine oxidase inhibitor iproniazid and the tricyclic antidepressant imipramine were effective antidepressants. Thus, the need for improved animal models to provide insights into the neuropathology underlying the disease is critical. Such information is in turn crucial for identifying new antidepressants and mood stabilisers. Currently, there is a shift away from traditional animal models to more focused research dealing with an endophenotype-style approach, genetic models, and incorporation of new findings from human neuroimaging and genetic studies. Such approaches are opening up more tractable avenues for understanding the neurobiological and genetic bases of these disorders. Further, such models promise to yield better translational animal models and hence more fruitful therapeutic targets. This overview focuses on such animal models and tests and how they can be used to assess MDD and BD in rodents.
Collapse
|
21
|
Sotnikov S, Wittmann A, Bunck M, Bauer S, Deussing J, Schmidt M, Touma C, Landgraf R, Czibere L. Blunted HPA axis reactivity reveals glucocorticoid system dysbalance in a mouse model of high anxiety-related behavior. Psychoneuroendocrinology 2014; 48:41-51. [PMID: 24995583 DOI: 10.1016/j.psyneuen.2014.06.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/19/2014] [Accepted: 06/10/2014] [Indexed: 01/15/2023]
Abstract
Depression and anxiety disorders are often characterized by altered hypothalamic-pituitary-adrenal (HPA) axis re-/activity. However, the presence of a molecular link between dysbalanced neuroendocrine regulation and psychopathologies is not yet fully established. Earlier, we reported that high (HAB), normal (NAB) and low (LAB) anxiety-related behavior mice express divergent anxiety-related and passive/active coping phenotypes. Here, we studied mechanisms that might contribute to the different HPA axis reactivity observed in HAB, NAB and LAB mice and their involvement in the regulation of anxiety-related behavior and passive/active coping style. We found that HAB mice respond with significantly reduced corticosterone (CORT) secretion to an acute stressful stimulus and a blunted response in the Dex/CRH test compared to NAB and LAB mice. At the molecular level, higher expression of the glucocorticoid receptor (GR/Nr3c1) and decreased corticotropin-releasing hormone receptor 1 (CRHR1) expression were observed in the pituitary of HAB mice. We further analyzed whether these stress mediators differed between the HAB, NAB and LAB lines in limbic system-associated brain regions and whether their interplay contributes to the phenotype. Interestingly, not only in the pituitary but also in almost all brain regions investigated, GR expression was significantly higher in HAB mice. In contrast, the amount of CORT in the brain structures analyzed was significantly lower in these animals. The expression of CRHR1 varied in the prefrontal cortex only. Since glucocorticoids regulate both GR and CRHR1, we treated HAB and NAB mice chronically with CORT. After 6 weeks of administration, reduced anxiety- and depression-like behaviors were observed in HAB mice, whereas increased anxiety was found in NABs. In both groups, GR, but not CRHR1, were significantly reduced. Taken together, our study proposes HAB mice as an animal model of simultaneous features of increased anxiety-related and depression-like behaviors with blunted HPA axis reactivity suggesting a dysregulated GR/CORT system as one key mechanism behind their phenotype.
Collapse
Affiliation(s)
- Sergey Sotnikov
- Max Planck Institute of Psychiatry, 80804 Munich, Germany; Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia.
| | - Anke Wittmann
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Mirjam Bunck
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Sabrina Bauer
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Jan Deussing
- Max Planck Institute of Psychiatry, 80804 Munich, Germany; Clinical Cooperation Group Molecular Neurogenetics, Institute of Developmental Genetics, Helmholtz Center Munich, 85764 Neuherberg, Germany
| | | | - Chadi Touma
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | | | - Ludwig Czibere
- Max Planck Institute of Psychiatry, 80804 Munich, Germany
| |
Collapse
|
22
|
He F. The relationship of prenatal ethanol exposure and anxiety-related behaviors and central androgen receptor and vasopressin expression in adult male mandarin voles. Neuroscience 2014; 266:224-34. [DOI: 10.1016/j.neuroscience.2014.02.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 02/14/2014] [Accepted: 02/15/2014] [Indexed: 01/13/2023]
|
23
|
Yan Y, Wang YL, Su Z, Zhang Y, Guo SX, Liu AJ, Wang CH, Sun FJ, Yang J. Effect of oxytocin on the behavioral activity in the behavioral despair depression rat model. Neuropeptides 2014; 48:83-9. [PMID: 24444823 DOI: 10.1016/j.npep.2014.01.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 11/23/2013] [Accepted: 01/01/2014] [Indexed: 02/02/2023]
Abstract
Oxytocin (OXT), a nonapeptide posterior hormone of the pituitary, is mainly synthesized and secreted in the hypothalamic paraventricular nucleus (PVN) and supraoptic nucleus (SON). The present study was to investigate in which level, brain or periphery, OXT effecting on the behavioral activity in the behavioral despair depression rat model. The results showed that (1) either the forced swimming or the tail suspension significantly increased OXT concentration in the brain (PVN, SON, frontal cortex, hippocampus, amygdala, lumbar spinal cord) and in the periphery (posterior pituitary and serum); (2) intraventricular injection (icv) of OXT decreased the animal immobility time, whereas OXT receptor antagonist-desGly-NH2, d(CH2)5[D-Tyr2, Thr-sup-4]OV (icv) increased the animal immobility time in a dose-dependent manner in forced swimming test (FST) and in tail suspension test (TST); (3) neither OXT nor OXT receptor antagonist (intravenous injection) influenced the animal immobility time in FST and in TST. OXT levels were increased in several areas of the brain and in the periphery following the behavioral despair, one stressor, yet pre-treatment with OXT appeared to be beneficial in term of reducing immobility time. The data suggested that behavioral despair could enhance OXT synthesis and secretion not only in the brain but also in the periphery, and OXT in the brain rather than the periphery played a role in the behavioral despair depression.
Collapse
Affiliation(s)
- Yuan Yan
- The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, China
| | - Yong-Liang Wang
- The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, China
| | - Zhou Su
- The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, China
| | - Yi Zhang
- The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, China
| | - Shuang-Xi Guo
- The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, China
| | - Ai-Juan Liu
- The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan 453100, China
| | - Chang-Hong Wang
- Henan Provincial Mental Hospital, Xinxiang, Henan 453002, China
| | - Fang-Jie Sun
- Xinxiang Institute for New Medicine, Xinxiang, Henan 453003, China
| | - Jun Yang
- Xinxiang Institute for New Medicine, Xinxiang, Henan 453003, China; Henan Fenghuang Pharmaceutical Co. Ltd., Xinxiang, Henan 453003, China.
| |
Collapse
|
24
|
Chekmareva NY, Sotnikov SV, Diepold RP, Naik RR, Landgraf R, Czibere L. Environmental manipulations generate bidirectional shifts in both behavior and gene regulation in a crossbred mouse model of extremes in trait anxiety. Front Behav Neurosci 2014; 8:87. [PMID: 24672450 PMCID: PMC3957683 DOI: 10.3389/fnbeh.2014.00087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 02/28/2014] [Indexed: 01/08/2023] Open
Abstract
Although gene-environment interactions are known to significantly influence psychopathology-related disease states, only few animal models cover both the genetic background and environmental manipulations. Therefore, we have taken advantage of the bidirectionally inbred high (HAB) and low (LAB) anxiety-related behavior mouse lines to generate HAB × LAB F1 hybrids that intrinsically carry both lines’ genetic characteristics, and subsequently raised them in three different environments—standard, enriched (EE) and chronic mild stress (CMS). Assessing genetic correlates of trait anxiety, we focused on two genes already known to play a role in HAB vs. LAB mice, corticotropin releasing hormone receptor type 1 (Crhr1) and high mobility group nucleosomal binding domain 3 (Hmgn3). While EE F1 mice showed decreased anxiety-related and increased explorative behaviors compared to controls, CMS sparked effects in the opposite direction. However, environmental treatments affected the expression of the two genes in distinct ways. Thus, while expression ratios of Hmgn3 between the HAB- and LAB-specific alleles remained equal, total expression resembled the one observed in HAB vs. LAB mice, i.e., decreased after EE and increased after CMS treatment. On the other hand, while total expression of Crhr1 remained unchanged between the groups, the relative expression of HAB- and LAB-specific alleles showed a clear effect following the environmental modifications. Thus, the environmentally driven bidirectional shift of trait anxiety in this F1 model strongly correlated with Hmgn3 expression, irrespective of allele-specific expression patterns that retained the proportions of basic differential HAB vs. LAB expression, making this gene a match for environment-induced modifications. An involvement of Crhr1 in the bidirectional behavioral shift could, however, rather be due to different effects of the HAB- and LAB-specific alleles described here. Both candidate genes therefore deserve attention in the complex regulation of anxiety-related phenotypes including environment-mediated effects.
Collapse
Affiliation(s)
- Natalia Yurievna Chekmareva
- Department of Behavioral Neuroendocrinology, Max Planck Institute of Psychiatry Munich, Germany ; Department of Normal Physiology, Sechenov First Moscow State Medical University Moscow, Russia
| | - Sergey V Sotnikov
- Department of Behavioral Neuroendocrinology, Max Planck Institute of Psychiatry Munich, Germany ; Department of Normal Physiology, Sechenov First Moscow State Medical University Moscow, Russia
| | - Rebekka P Diepold
- Department of Behavioral Neuroendocrinology, Max Planck Institute of Psychiatry Munich, Germany
| | - Roshan R Naik
- Department of Behavioral Neuroendocrinology, Max Planck Institute of Psychiatry Munich, Germany
| | - Rainer Landgraf
- Department of Behavioral Neuroendocrinology, Max Planck Institute of Psychiatry Munich, Germany
| | - Ludwig Czibere
- Department of Behavioral Neuroendocrinology, Max Planck Institute of Psychiatry Munich, Germany
| |
Collapse
|
25
|
Yen YC, Anderzhanova E, Bunck M, Schuller J, Landgraf R, Wotjak CT. Co-segregation of hyperactivity, active coping styles, and cognitive dysfunction in mice selectively bred for low levels of anxiety. Front Behav Neurosci 2013; 7:103. [PMID: 23966915 PMCID: PMC3744008 DOI: 10.3389/fnbeh.2013.00103] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 07/26/2013] [Indexed: 11/13/2022] Open
Abstract
We established mouse models of extremes in trait anxiety, which are based on selective breeding for low vs. normal vs. high open-arm exploration on the elevated plus-maze. Genetically selected low anxiety-related behavior (LAB) coincided with hyperactivity in the home cage. Given the fact that several psychiatric disorders such as schizophrenia, mania, and attention deficit hyperactivity disorder (ADHD) share hyperactivity symptom, we systematically examined LAB mice with respect to unique and overlapping endophenotypes of the three diseases. To this end Venn diagrams were used as an instrument for discrimination of possible models. We arranged the endophenotypes in Venn diagrams and translated them into different behavioral tests. LAB mice showed elevated levels of locomotion in the open field (OF) test with deficits in habituation, compared to mice bred for normal (NAB) and high anxiety-related behavior (HAB). Cross-breeding of hypoactive HAB and hyperactive LAB mice resulted in offspring showing a low level of locomotion comparable to HAB mice, indicating that the HAB alleles are dominant over LAB alleles in determining the level of locomotion. In a holeboard test, LAB mice spent less time in hole exploration, as shown in patients with schizophrenia and ADHD; however, LAB mice displayed no impairments in social interaction and prepulse inhibition (PPI), implying a unlikelihood of LAB as an animal model of schizophrenia. Although LAB mice displayed hyperarousal, active coping styles, and cognitive deficits, symptoms shared by mania and ADHD, they failed to reveal the classic manic endophenotypes, such as increased hedonia and object interaction. The neuroleptic haloperidol reduced locomotor activity in all mouse lines. The mood stabilizer lithium and the psychostimulant amphetamine, in contrast, selectively reduced hyperactivity in LAB mice. Based on the behavioral and pharmacological profiles, LAB mice are suggested as a novel rodent model of ADHD-like symptoms.
Collapse
Affiliation(s)
- Yi-Chun Yen
- Department of Neuronal Plasticity, Max Planck Institute of Psychiatry Munich, Germany
| | | | | | | | | | | |
Collapse
|
26
|
Fabio KM, Guillon CD, Lu SF, Heindel ND, Brownstein MJ, Lacey CJ, Garippa C, Simon NG. Pharmacokinetics and Metabolism of SRX246: A Potent and Selective Vasopressin 1a Antagonist. J Pharm Sci 2013; 102:2033-2043. [DOI: 10.1002/jps.23495] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 02/11/2013] [Accepted: 02/12/2013] [Indexed: 02/05/2023]
|
27
|
Identification of a role for the ventral hippocampus in neuropeptide S-elicited anxiolysis. PLoS One 2013; 8:e60219. [PMID: 23555930 PMCID: PMC3610821 DOI: 10.1371/journal.pone.0060219] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Accepted: 02/21/2013] [Indexed: 12/15/2022] Open
Abstract
Neuropeptide S (NPS) increasingly emerges as a potential novel treatment option for anxiety diseases like panic and posttraumatic stress disorder. However, the neural underpinnings of its anxiolytic action are still not clearly understood. Recently, we reported that neurons of the ventral hippocampus (VH) take up intranasally administered fluorophore-conjugated NPS and, moreover, that application of NPS to mouse brain slices affects neurotransmission and plasticity at hippocampal CA3-CA1 synapses. Although these previous findings define the VH as a novel NPS target structure, they leave open whether this brain region is directly involved in NPS-mediated anxiolysis and how NPS impacts on neuronal activity propagation in the VH. Here, we fill this knowledge gap by demonstrating, first, that microinjections of NPS into the ventral CA1 region are sufficient to reduce anxiety-like behavior of C57BL/6N mice and, second, that NPS, via the NPS receptor, rapidly weakens evoked neuronal activity flow from the dentate gyrus to area CA1 in vitro. Additionally, we show that intranasally applied NPS alters neurotransmission and plasticity at CA3-CA1 synapses in the same way as NPS administered to hippocampal slices. Thus, our study provides, for the first time, strong experimental evidence for a direct involvement of the VH in NPS-induced anxiolysis and furthermore presents a novel mechanism of NPS action.
Collapse
|
28
|
Distler MG, Palmer AA. Role of Glyoxalase 1 (Glo1) and methylglyoxal (MG) in behavior: recent advances and mechanistic insights. Front Genet 2012. [PMID: 23181072 PMCID: PMC3500958 DOI: 10.3389/fgene.2012.00250] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glyoxalase 1 (GLO1) is a ubiquitous cellular enzyme that participates in the detoxification of methylglyoxal (MG), a cytotoxic byproduct of glycolysis that induces protein modification (advanced glycation end-products, AGEs), oxidative stress, and apoptosis. The concentration of MG is elevated under high-glucose conditions, such as diabetes. As such, GLO1 and MG have been implicated in the pathogenesis of diabetic complications. Recently, findings have linked GLO1 to numerous behavioral phenotypes, including psychiatric diseases (anxiety, depression, schizophrenia, and autism) and pain. This review highlights GLO1's association with behavioral phenotypes, describes recent discoveries that have elucidated the underlying mechanisms, and identifies opportunities for future research.
Collapse
|
29
|
Gonik M, Frank E, Keßler MS, Czamara D, Bunck M, Yen YC, Pütz B, Holsboer F, Bettecken T, Landgraf R, Müller-Myhsok B, Touma C, Czibere L. The endocrine stress response is linked to one specific locus on chromosome 3 in a mouse model based on extremes in trait anxiety. BMC Genomics 2012; 13:579. [PMID: 23114097 PMCID: PMC3557225 DOI: 10.1186/1471-2164-13-579] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 10/29/2012] [Indexed: 12/17/2022] Open
Abstract
Background The hypothalamic-pituitary-adrenal (HPA) axis is essential to control physiological stress responses in mammals. Its dysfunction is related to several mental disorders, including anxiety and depression. The aim of this study was to identify genetic loci underlying the endocrine regulation of the HPA axis. Method High (HAB) and low (LAB) anxiety-related behaviour mice were established by selective inbreeding of outbred CD-1 mice to model extremes in trait anxiety. Additionally, HAB vs. LAB mice exhibit comorbid characteristics including a differential corticosterone response upon stress exposure. We crossbred HAB and LAB lines to create F1 and F2 offspring. To identify the contribution of the endocrine phenotypes to the total phenotypic variance, we examined multiple behavioural paradigms together with corticosterone secretion-based phenotypes in F2 mice by principal component analysis. Further, to pinpoint the genomic loci of the quantitative trait of the HPA axis stress response, we conducted genome-wide multipoint oligogenic linkage analyses based on Bayesian Markov chain Monte Carlo approach as well as parametric linkage in three-generation pedigrees, followed by a two-dimensional scan for epistasis and association analysis in freely segregating F2 mice using 267 single-nucleotide polymorphisms (SNPs), which were identified to consistently differ between HAB and LAB mice as genetic markers. Results HPA axis reactivity measurements and behavioural phenotypes were represented by independent principal components and demonstrated no correlation. Based on this finding, we identified one single quantitative trait locus (QTL) on chromosome 3 showing a very strong evidence for linkage (2ln (L-score) > 10, LOD > 23) and significant association (lowest Bonferroni adjusted p < 10-28) to the neuroendocrine stress response. The location of the linkage peak was estimated at 42.3 cM (95% confidence interval: 41.3 - 43.3 cM) and was shown to be in epistasis (p-adjusted < 0.004) with the locus at 35.3 cM on the same chromosome. The QTL harbours genes involved in steroid synthesis and cardiovascular effects. Conclusion The very prominent effect on stress-induced corticosterone secretion of the genomic locus on chromosome 3 and its involvement in epistasis highlights the critical role of this specific locus in the regulation of the HPA axis.
Collapse
Affiliation(s)
- Mariya Gonik
- Max Planck Institute of Psychiatry, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Haller J. The neurobiology of abnormal manifestations of aggression--a review of hypothalamic mechanisms in cats, rodents, and humans. Brain Res Bull 2012; 93:97-109. [PMID: 23085544 DOI: 10.1016/j.brainresbull.2012.10.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 10/08/2012] [Accepted: 10/09/2012] [Indexed: 01/01/2023]
Abstract
Aggression research was for long dominated by the assumption that aggression-related psychopathologies result from the excessive activation of aggression-promoting brain mechanisms. This assumption was recently challenged by findings with models of aggression that mimic etiological factors of aggression-related psychopathologies. Subjects submitted to such procedures show abnormal attack features (mismatch between provocation and response, disregard of species-specific rules, and insensitivity toward the social signals of opponents). We review here 12 such laboratory models and the available human findings on the neural background of abnormal aggression. We focus on the hypothalamus, a region tightly involved in the execution of attacks. Data show that the hypothalamic mechanisms controlling attacks (general activation levels, local serotonin, vasopressin, substance P, glutamate, GABA, and dopamine neurotransmission) undergo etiological factor-dependent changes. Findings suggest that the emotional component of attacks differentiates two basic types of hypothalamic mechanisms. Aggression associated with increased arousal (emotional/reactive aggression) is paralleled by increased mediobasal hypothalamic activation, increased hypothalamic vasopressinergic, but diminished hypothalamic serotonergic neurotransmission. In aggression models associated with low arousal (unemotional/proactive aggression), the lateral but not the mediobasal hypothalamus is over-activated. In addition, the anti-aggressive effect of serotonergic neurotransmission is lost and paradoxical changes were noticed in vasopressinergic neurotransmission. We conclude that there is no single 'neurobiological road' to abnormal aggression: the neural background shows qualitative, etiological factor-dependent differences. Findings obtained with different models should be viewed as alternative mechanisms rather than conflicting data. The relevance of these findings for understanding and treating of aggression-related psychopathologies is discussed. This article is part of a Special Issue entitled 'Extrasynaptic ionotropic receptors'.
Collapse
Affiliation(s)
- Jozsef Haller
- Department of Behavioral Neurobiology, Hungarian Academy of Sciences, Institute of Experimental Medicine, H-1083 Budapest, Szigony utca 43, Hungary.
| |
Collapse
|
31
|
Sah A, Schmuckermair C, Sartori SB, Gaburro S, Kandasamy M, Irschick R, Klimaschewski L, Landgraf R, Aigner L, Singewald N. Anxiety- rather than depression-like behavior is associated with adult neurogenesis in a female mouse model of higher trait anxiety- and comorbid depression-like behavior. Transl Psychiatry 2012; 2:e171. [PMID: 23047242 PMCID: PMC3565824 DOI: 10.1038/tp.2012.94] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 08/22/2012] [Accepted: 08/22/2012] [Indexed: 02/07/2023] Open
Abstract
Adult neurogenesis has been implicated in affective disorders and the action of antidepressants (ADs) although the functional significance of this association is still unclear. The use of animal models closely mimicking human comorbid affective and anxiety disorders seen in the majority of patients should provide relevant novel information. Here, we used a unique genetic mouse model displaying higher trait anxiety (HAB) and comorbid depression-like behavior. We demonstrate that HABs have a lower rate of hippocampal neurogenesis and impaired functional integration of newly born neurons as compared with their normal anxiety/depression-like behavior (NAB) controls. In HABs, chronic treatment with the AD fluoxetine alleviated their higher depression-like behavior and protected them from relapse for 3 but not 7 weeks after discontinuation of the treatment without affecting neurogenesis. Similar to what has been observed in depressed patients, fluoxetine treatment induced anxiogenic-like effects during the early treatment phase in NABs along with a reduction in neurogenesis. On the other hand, treatment with AD drugs with a particularly strong anxiolytic component, namely the neurokinin-1-receptor-antagonist L-822 429 or tianeptine, increased the reduced rate of neurogenesis in HABs up to NAB levels. In addition, challenge-induced hypoactivation of dentate gyrus (DG) neurons in HABs was normalized by all three drugs. Overall, these data suggest that AD-like effects in a psychopathological mouse model are commonly associated with modulation of DG hypoactivity but not neurogenesis, suggesting normalization of hippocampal hypoactivity as a neurobiological marker indicating successful remission. Finally, rather than to higher depression-related behavior, neurogenesis seems to be linked to pathological anxiety.
Collapse
Affiliation(s)
- A Sah
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Centre for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
- Max-Planck-Institute of Psychiatry, Kraepelinstrasse, Munich,Germany
| | - C Schmuckermair
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Centre for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - S B Sartori
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Centre for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - S Gaburro
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Centre for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - M Kandasamy
- Institut für Molekulare Regenerative Medizin, Paracelsus Medizinische Privatuniversität, Strubergasse, Salzburg, Austria
| | - R Irschick
- Division of Neuroanatomy, Department of Anatomy, Histology and Embryology, Innsbruck, Austria
| | - L Klimaschewski
- Division of Neuroanatomy, Department of Anatomy, Histology and Embryology, Innsbruck, Austria
| | - R Landgraf
- Max-Planck-Institute of Psychiatry, Kraepelinstrasse, Munich,Germany
| | - L Aigner
- Institut für Molekulare Regenerative Medizin, Paracelsus Medizinische Privatuniversität, Strubergasse, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg, Paracelsus Medical University, Strubergasse, Salzburg, Austria
| | - N Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Centre for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
32
|
Neumann ID, Landgraf R. Balance of brain oxytocin and vasopressin: implications for anxiety, depression, and social behaviors. Trends Neurosci 2012; 35:649-59. [PMID: 22974560 DOI: 10.1016/j.tins.2012.08.004] [Citation(s) in RCA: 680] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Revised: 05/24/2012] [Accepted: 08/13/2012] [Indexed: 02/07/2023]
Abstract
Oxytocin and vasopressin are regulators of anxiety, stress-coping, and sociality. They are released within hypothalamic and limbic areas from dendrites, axons, and perikarya independently of, or coordinated with, secretion from neurohypophysial terminals. Central oxytocin exerts anxiolytic and antidepressive effects, whereas vasopressin tends to show anxiogenic and depressive actions. Evidence from pharmacological and genetic association studies confirms their involvement in individual variation of emotional traits extending to psychopathology. Based on their opposing effects on emotional behaviors, we propose that a balanced activity of both brain neuropeptide systems is important for appropriate emotional behaviors. Shifting the balance between the neuropeptide systems towards oxytocin, by positive social stimuli and/or psychopharmacotherapy, may help to improve emotional behaviors and reinstate mental health.
Collapse
Affiliation(s)
- Inga D Neumann
- Department of Behavioral and Molecular Neurobiology, University of Regensburg, Regensburg, Germany.
| | | |
Collapse
|
33
|
Yang J, Pan YJ, Yin ZK, Hai GF, Lu L, Zhao Y, Wang DX, Wang H, Wang G. Effect of arginine vasopressin on the behavioral activity in the behavior despair depression rat model. Neuropeptides 2012; 46:141-9. [PMID: 22513399 DOI: 10.1016/j.npep.2012.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 03/24/2012] [Accepted: 03/26/2012] [Indexed: 02/03/2023]
Abstract
Arginine vasopressin (AVP), a nonapeptide posterior hormone of the pituitary, is mainly synthesized and secreted in the hypothalamic paraventricular nucleus (PVN) and supraoptic nucleus (SON). Large numbers of studies have reported that AVP plays a role in depression. The present study was to investigate by which level, brain or periphery, AVP affects the behavioral activity in the behavior despair depression rat model. The results showed that (1) either forced swimming or tail suspension significantly increased AVP concentration not only in the brain (PVN, SON, frontal of cortex, hippocampus, amygdala, lumber spinal cord) but also in the periphery (posterior pituitary and serum); (2) intraventricular injection (icv) of AVP decreased the animal immobility time, whereas V₁ receptor antagonist d(CH₂)₅Tyr(Me)AVP (icv) increased the animal immobility time in a dose-dependent manner not only in FST but also in TST, but the V₂ receptor antagonist d(CH₂)₅[D-Ile, Ile, Ala-NH₉]AVP did not change the animal immobility time in FST or TST; (3) V₁, not V₂ receptor antagonist could inhibit the animal immobility time decrease induced by AVP (icv); (4) neither AVP nor its receptor antagonist (including V₁ and V₂ receptor antagonist) influenced the animal immobility time in both FST and TST. The data suggested that AVP in the brain rather than the periphery played a role in the behavior despair depression by V₁, not V₂ receptors, which behavior despair might have a positive feedback effect on central AVP and blood AVP might have a negative feedback on central AVP in the depressive process.
Collapse
Affiliation(s)
- Jun Yang
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan 453003, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Jurek B, Slattery DA, Maloumby R, Hillerer K, Koszinowski S, Neumann ID, van den Burg EH. Differential contribution of hypothalamic MAPK activity to anxiety-like behaviour in virgin and lactating rats. PLoS One 2012; 7:e37060. [PMID: 22615888 PMCID: PMC3355176 DOI: 10.1371/journal.pone.0037060] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 04/12/2012] [Indexed: 12/18/2022] Open
Abstract
The c-Raf – MEK1/2 – ERK1/2 mitogen-activated protein kinase (MAPK) intracellular signalling cascade in neurons plays important roles in the control of a variety of behaviours, including social behaviours and anxiety. These roles partially overlap with those described for oxytocin (OXT), and it has been shown that OXT activates the MAPK pathway in the hypothalamus (of male), and hippocampus (of female) rats. Here, by combining behavioural (light/dark box) and biochemical analyses (western blotting), we tested two hypotheses: (i) that OXT is anxiolytic within the hypothalamus of females, and (ii) that this effect, as well as that of lactation-associated anxiolysis, depends on the recruitment of the MAPK pathway. We found that, when injected bilaterally into the hypothalamic paraventricular nucleus (PVN), OXT decreased anxiety-like behaviour in virgins, and that this effect depended on phosphorylation of MEK1/2. MAPK pathway activation in lactation was evident by high phosphorylated (p) MEK1/2 levels, and nuclear translocation of ERK1. The high pMEK1/2 levels were necessary for the anxiolytic phenotype typically observed during lactation. Interestingly, exogenous OXT in lactating rats reduced pMEK1/2 levels without a concomitant effect on anxiety, indicating that OXT receptor activation can lead to recruitment of additional intracellular pathways to modulate MEK activity. Still other pathways could include MEK, but without subsequent activation of ERK, as we did not observe any increase in OXT-induced ERK phosphorylation. Together the results demonstrate that the MAPK pathway, especially MEK1/2, is critically involved in the regulation of anxiety-like behaviour in female rats.
Collapse
Affiliation(s)
- Benjamin Jurek
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| | - David A. Slattery
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| | - Rodrigue Maloumby
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| | - Katharina Hillerer
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| | - Sophie Koszinowski
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| | - Inga D. Neumann
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
- * E-mail: Inga
| | - Erwin H. van den Burg
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
35
|
Intranasally administered neuropeptide S (NPS) exerts anxiolytic effects following internalization into NPS receptor-expressing neurons. Neuropsychopharmacology 2012; 37:1323-37. [PMID: 22278093 PMCID: PMC3327839 DOI: 10.1038/npp.2011.317] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Experiments in rodents revealed neuropeptide S (NPS) to constitute a potential novel treatment option for anxiety diseases such as panic and post-traumatic stress disorder. However, both its cerebral target sites and the molecular underpinnings of NPS-mediated effects still remain elusive. By administration of fluorophore-conjugated NPS, we pinpointed NPS target neurons in distinct regions throughout the entire brain. We demonstrated their functional relevance in the hippocampus. In the CA1 region, NPS modulates synaptic transmission and plasticity. NPS is taken up into NPS receptor-expressing neurons by internalization of the receptor-ligand complex as we confirmed by subsequent cell culture studies. Furthermore, we tracked internalization of intranasally applied NPS at the single-neuron level and additionally demonstrate that it is delivered into the mouse brain without losing its anxiolytic properties. Finally, we show that NPS differentially modulates the expression of proteins of the glutamatergic system involved inter alia in synaptic plasticity. These results not only enlighten the path of NPS in the brain, but also establish a non-invasive method for NPS administration in mice, thus strongly encouraging translation into a novel therapeutic approach for pathological anxiety in humans.
Collapse
|
36
|
Wegener G, Finger BC, Elfving B, Keller K, Liebenberg N, Fischer CW, Singewald N, Slattery DA, Neumann ID, Mathé AA. Neuropeptide S alters anxiety, but not depression-like behaviour in Flinders Sensitive Line rats: a genetic animal model of depression. Int J Neuropsychopharmacol 2012; 15:375-87. [PMID: 21708052 DOI: 10.1017/s1461145711000678] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Neuropeptide S (NPS) and its receptor (NPSR) have been implicated in the mediation of anxiolytic-like behaviour in rodents. However, little knowledge is available regarding the NPS system in depression-related behaviours, and whether NPS also exerts anxiolytic effects in an animal model of psychopathology. Therefore, the aim of this work was to characterize the effects of NPS on depression- and anxiety-related parameters, using male and female rats in a well-validated animal model of depression: the Flinders Sensitive Line (FSL), their controls, the Flinders Resistant Line (FRL), and Sprague-Dawley (SD) rats. We found that FSL showed greater immobility in the forced swim test (FST) than FRL, confirming their phenotype. However, NPS did not affect depression-related behaviour in any rat line. No significant differences in baseline anxiety levels between the FSL and FRL strains were observed, but FSL and FRL rats displayed less anxiety-like behaviour compared to SD rats. NPS decreased anxiety-like behaviour on the elevated plus-maze in all strains. The expression of the NPSR in the amygdala, periventricular hypothalamic nucleus, and hippocampus was equal in all male strains, although a trend towards reduced expression within the amygdala was observed in FSL rats compared to SD rats. In conclusion, NPS had a marked anxiolytic effect in FSL, FRL and SD rats, but did not modify the depression-related behaviour in any strain, in spite of the significant differences in innate level between the strains. These findings suggest that NPS specifically modifies anxiety behaviour but cannot overcome/reverse a genetically mediated depression phenotype.
Collapse
Affiliation(s)
- Gregers Wegener
- Centre for Psychiatric Research, Aarhus University, Aarhus, Denmark
| | - Beate C Finger
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Betina Elfving
- Centre for Psychiatric Research, Aarhus University, Aarhus, Denmark
| | - Kirsten Keller
- Centre for Psychiatric Research, Aarhus University, Aarhus, Denmark
| | - Nico Liebenberg
- Centre for Psychiatric Research, Aarhus University, Aarhus, Denmark
| | | | - Nicolas Singewald
- Department of Pharmacology and Toxicology & Centre of Molecular Biosciences Innsbruck, University of Innsbruck, Austria
| | - David A Slattery
- Department of Neurobiology, Institute of Zoology, University of Regensburg, Regensburg, Germany
| | - Inga D Neumann
- Department of Neurobiology, Institute of Zoology, University of Regensburg, Regensburg, Germany
| | - Aleksander A Mathé
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
37
|
Abstract
Stress has been identified as a key risk factor for a multitude of human pathologies. However, stress by itself is often not sufficient to induce a disease, as a large contribution comes from an individual's genetic background. Therefore, many stress models have been created to investigate this so-called gene-environment interaction for different diseases. Recently, evidence has been accumulating to indicate that not only the exposure to stress, but also the vulnerability to such an exposure can have a significant impact on the development of disease. Herein we review recent animal models of stress vulnerability and resilience, with special attention devoted to the readout parameters and the potential for translatability of the results.
Collapse
Affiliation(s)
- Sebastian H Scharf
- Max Planck Institute of Psychiatry, Kraepelinstr. 2, 80804, Munich, Germany.
| | | |
Collapse
|
38
|
Wegener G, Mathe AA, Neumann ID. Selectively bred rodents as models of depression and anxiety. Curr Top Behav Neurosci 2012; 12:139-187. [PMID: 22351423 DOI: 10.1007/7854_2011_192] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Stress related diseases such as depression and anxiety have a high degree of co morbidity, and represent one of the greatest therapeutic challenges for the twenty-first century. The present chapter will summarize existing rodent models for research in psychiatry, mimicking depression- and anxiety-related diseases. In particular we will highlight the use of selective breeding of rodents for extremes in stress-related behavior. We will summarize major behavioral, neuroendocrine and neuronal parameters, and pharmacological interventions, assessed in great detail in two rat model systems: The Flinders Sensitive and Flinders Resistant Line rats (FSL/FRL model), and rats selectively bred for high (HAB) or low (LAB) anxiety related behavior (HAB/LAB model). Selectively bred rodents also provide an excellent tool in order to study gene and environment interactions. Although it is generally accepted that genes and environmental factors determine the etiology of mental disorders, precise information is limited: How rigid is the genetic disposition? How do genetic, prenatal and postnatal influences interact to shape adult disease? Does the genetic predisposition determine the vulnerability to prenatal and postnatal or adult stressors? In combination with modern neurobiological methods, these models are important to elucidate the etiology and pathophysiology of anxiety and affective disorders, and to assist in the development of new treatment paradigms.
Collapse
Affiliation(s)
- Gregers Wegener
- Centre for Psychiatric Research, Aarhus University Hospital, 8240, Risskov, Denmark,
| | | | | |
Collapse
|
39
|
Fabio K, Guillon C, Lacey CJ, Lu SF, Heindel ND, Ferris CF, Placzek M, Jones G, Brownstein MJ, Simon NG. Synthesis and evaluation of potent and selective human V1a receptor antagonists as potential ligands for PET or SPECT imaging. Bioorg Med Chem 2011; 20:1337-45. [PMID: 22249122 DOI: 10.1016/j.bmc.2011.12.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 12/05/2011] [Accepted: 12/07/2011] [Indexed: 01/17/2023]
Abstract
SRX246 is a potent, highly selective human vasopressin V1a antagonist that crosses the blood-brain barrier in rats. CNS penetration makes SRX246 an ideal candidate for potential radiolabeling and use in visualization and characterization of the role of the V1a receptor in multiple stress-related disorders. Before radiolabeling studies, cold reference analogs of SRX246 were prepared. This study describes the synthesis and in vitro screening for human V1a receptor binding and permeability of fluoro, iodo, and methyl reference compounds for SRX246 and the preparation of a tin precursor. For each compound, the potential utility of corresponding radiolabeled analogs for PET and SPECT imaging is discussed.
Collapse
Affiliation(s)
- Karine Fabio
- Dept. of Chemistry, Lehigh University, Bethlehem, PA 18015, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Russell R, Doyle R, Turner J, Attkins N, Ramsey S, Weibley L, Bateman L, Bictash M, Neal-Morgan S, Ivarsson M, Pullen N. In vitro and in vivo pharmacological characterisation of the potent and selective vasopressin V(1A) receptor antagonist 4-[4-(4-Chloro-phenyl)-5-[1,2,3]triazol-2-ylmethyl-4H-[1,2,4]triazol-3-yl]-piperidin-1-yl-(3,5-difluoro-phenyl) methanone (PF-00738245). Eur J Pharmacol 2011; 670:347-55. [PMID: 21958878 DOI: 10.1016/j.ejphar.2011.09.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 09/05/2011] [Accepted: 09/11/2011] [Indexed: 10/17/2022]
Abstract
The dysregulation of arginine vasopressin (AVP) release and activation of vasopressin receptors plays an important role in disease conditions including polycystic kidney disease, congestive heart failure and dysmenorrhoea. The development of potent and selective vasopressin receptor ligands is needed to help dissect the function of the specific subtypes in disease pathogenesis. Here we report the pharmacological characterisation of PF-00738245 in in vitro binding and functional assays using cells expressing vasopressin V(₁A), V(₁B) or V₂ receptors. PF-00738245 inhibited AVP binding to the recombinant human vasopressin V(₁A) receptor (K(i)=2.85 nM) and blocked AVP-induced rat aortic ring and human myometrial contraction (pK(B)=7.35 and 8.62 respectively). PF-00738245 was selective for the vasopressin V(1A) receptor by demonstrating minimal binding to vasopressin V(₁B) (3.6% inhibition at 10 μM) or functional activity at vasopressin V₂ receptors (8.1% agonist and -8.4% antagonist activity at 10 μM) as well as the oxytocin receptor (46.3% antagonist activity at 10 μM). The in vivo pharmacological properties were tested orally in the rat and PF-00738245 dose dependently blocked the effect of AVP on a capsaicin-induced cutaneous flare response. Taken together the data support the use of PF-00738245 as a potent and selective vasopressin V(₁A) receptor antagonist which may have utility in the treatment of disease conditions which are propagated by elevation in vasopressin V(₁A) receptor signalling.
Collapse
Affiliation(s)
- Rachel Russell
- Pfizer Global Research & Development, Ramsgate Road, Sandwich, Kent CT13 9NJ, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Profiling trait anxiety: transcriptome analysis reveals cathepsin B (Ctsb) as a novel candidate gene for emotionality in mice. PLoS One 2011; 6:e23604. [PMID: 21897848 PMCID: PMC3163650 DOI: 10.1371/journal.pone.0023604] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2011] [Accepted: 07/20/2011] [Indexed: 11/19/2022] Open
Abstract
Behavioral endophenotypes are determined by a multitude of counteracting but precisely balanced molecular and physiological mechanisms. In this study, we aim to identify potential novel molecular targets that contribute to the multigenic trait “anxiety”. We used microarrays to investigate the gene expression profiles of different brain regions within the limbic system of mice which were selectively bred for either high (HAB) or low (LAB) anxiety-related behavior, and also show signs of comorbid depression-like behavior. We identified and confirmed sex-independent differences in the basal expression of 13 candidate genes, using tissue from the entire brain, including coronin 7 (Coro7), cathepsin B (Ctsb), muscleblind-like 1 (Mbnl1), metallothionein 1 (Mt1), solute carrier family 25 member 17 (Slc25a17), tribbles homolog 2 (Trib2), zinc finger protein 672 (Zfp672), syntaxin 3 (Stx3), ATP-binding cassette, sub-family A member 2 (Abca2), ectonucleotide pyrophosphatase/phosphodiesterase 5 (Enpp5), high mobility group nucleosomal binding domain 3 (Hmgn3) and pyruvate dehydrogenase beta (Pdhb). Additionally, we confirmed brain region-specific differences in the expression of synaptotagmin 4 (Syt4). Our identification of about 90 polymorphisms in Ctsb suggested that this gene might play a critical role in shaping our mouse model's behavioral endophenotypes. Indeed, the assessment of anxiety-related and depression-like behaviors of Ctsb knock-out mice revealed an increase in depression-like behavior in females. Altogether, our results suggest that Ctsb has significant effects on emotionality, irrespective of the tested mouse strain, making it a promising target for future pharmacotherapy.
Collapse
|
42
|
Sotnikov SV, Markt PO, Umriukhin AE, Landgraf R. Genetic predisposition to anxiety-related behavior predicts predator odor response. Behav Brain Res 2011; 225:230-4. [PMID: 21801755 DOI: 10.1016/j.bbr.2011.07.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Revised: 07/10/2011] [Accepted: 07/13/2011] [Indexed: 12/11/2022]
Abstract
While rodents have a keen sense of smell and largely depend on olfactory cues for operating in their environment, most of the widely used tests to assess anxiety-related behavior largely ignore the olfactory system, being primarily based on fear of brightly lit, novel and open spaces. Here, we aimed at testing whether the genetic predisposition to anxiety predicts the predator odor response in mice. In the first experiment, using the 3-chamber avoidance test in CD-1 mice, trimethylthiazoline (TMT), a synthetic fox fecal odor, was shown to induce stronger behavioral and neuroendocrine effects than cat odor and butyric acid, respectively, and was therefore chosen as aversive odor for the following series of experiments. In this series, bidirectionally, selectively inbred CD-1 mice with either high (HAB), intermediate (NAB) or low (LAB) anxiety-related behavior responded differently to TMT, with HABs spending significantly less time than both NABs and LABs in the chamber harbouring the predator odor. Importantly, this result is not confounded by any deficit of the olfactory system, as LAB and NAB mice, while not or only moderately responding to TMT, responded to both the pleasant odor of female urine and the repugnant odor of butyric acid. Probably due to the strength of TMT, a similar increase in corticosterone levels upon predator odor exposure was observed in all three groups. Together, the results suggest that, dependent on the genetic predisposition to extremes in anxiety-related behavior, mice differentially interpret the odor of a potential predator, making this type of avoidance behavior highly predictable.
Collapse
Affiliation(s)
- S V Sotnikov
- Max Planck Institute of Psychiatry, Kraepelinstr, 2, 80804 Munich, Germany.
| | | | | | | |
Collapse
|
43
|
Abstract
The lifetime prevalence of panic disorder (PD) is up to 4% worldwide and there is substantial evidence that genetic factors contribute to the development of PD. Single-nucleotide polymorphisms (SNPs) in TMEM132D, identified in a whole-genome association study (GWAS), were found to be associated with PD in three independent samples, with a two-SNP haplotype associated in each of three samples in the same direction, and with a P-value of 1.2e-7 in the combined sample (909 cases and 915 controls). Independent SNPs in this gene were also associated with the severity of anxiety symptoms in patients affected by PD or panic attacks as well as in patients suffering from unipolar depression. Risk genotypes for PD were associated with higher TMEM132D mRNA expression levels in the frontal cortex. In parallel, using a mouse model of extremes in trait anxiety, we could further show that anxiety-related behavior was positively correlated with Tmem132d mRNA expression in the anterior cingulate cortex, central to the processing of anxiety/fear-related stimuli, and that in this animal model a Tmem132d SNP is associated with anxiety-related behavior in an F2 panel. TMEM132D may thus be an important new candidate gene for PD as well as more generally for anxiety-related behavior.
Collapse
|
44
|
Kessler MS, Bosch OJ, Bunck M, Landgraf R, Neumann ID. Maternal care differs in mice bred for high vs. low trait anxiety: Impact of brain vasopressin and cross-fostering. Soc Neurosci 2011; 6:156-68. [DOI: 10.1080/17470919.2010.495567] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
45
|
Sartori SB, Hauschild M, Bunck M, Gaburro S, Landgraf R, Singewald N. Enhanced fear expression in a psychopathological mouse model of trait anxiety: pharmacological interventions. PLoS One 2011; 6:e16849. [PMID: 21386891 PMCID: PMC3046120 DOI: 10.1371/journal.pone.0016849] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Accepted: 01/14/2011] [Indexed: 01/23/2023] Open
Abstract
The propensity to develop an anxiety disorder is thought to be determined by genetic and environmental factors. Here we investigated the relationship between a genetic predisposition to trait anxiety and experience-based learned fear in a psychopathological mouse model. Male CD-1 mice selectively bred for either high (HAB), or normal (NAB) anxiety-related behaviour on the elevated plus maze were subjected to classical fear conditioning. During conditioning both mouse lines showed increased fear responses as assessed by freezing behaviour. However, 24 h later, HAB mice displayed more pronounced conditioned responses to both a contextual or cued stimulus when compared with NAB mice. Interestingly, 6 h and already 1 h after fear conditioning, freezing levels were high in HAB mice but not in NAB mice. These results suggest that trait anxiety determines stronger fear memory and/or a weaker ability to inhibit fear responses in the HAB line. The enhanced fear response of HAB mice was attenuated by treatment with either the α(2,3,5)-subunit selective benzodiazepine partial agonist L-838,417, corticosterone or the selective neurokinin-1 receptor antagonist L-822,429. Overall, the HAB mouse line may represent an interesting model (i) for identifying biological factors underlying misguided conditioned fear responses and (ii) for studying novel anxiolytic pharmacotherapies for patients with fear-associated disorders, including post-traumatic stress disorder and phobias.
Collapse
Affiliation(s)
- Simone B Sartori
- Department of Pharmacology and Toxicology, Center for Molecular Biosciences Innsbruck and Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria.
| | | | | | | | | | | |
Collapse
|
46
|
Abstract
Comprehensive clinical studies show that adverse conditions in early life can severely impact the developing brain and increase vulnerability to mood disorders later in life. During early postnatal life the brain exhibits high plasticity which allows environmental signals to alter the trajectories of rapidly developing circuits. Adversity in early life is able to shape the experience-dependent maturation of stress-regulating pathways underlying emotional functions and endocrine responses to stress, such as the hypothalamo-pituitary-adrenal (HPA) system, leading to long-lasting altered stress responsivity during adulthood. To date, the study of gene-environment interactions in the human population has been dominated by epidemiology. However, recent research in the neuroscience field is now advancing clinical studies by addressing specifically the mechanisms by which gene-environment interactions can predispose individuals toward psychopathology. To this end, appropriate animal models are being developed in which early environmental factors can be manipulated in a controlled manner. Here we will review recent studies performed with the common aim of understanding the effects of the early environment in shaping brain development and discuss the newly developing role of epigenetic mechanisms in translating early life conditions into long-lasting changes in gene expression underpinning brain functions. Particularly, we argue that epigenetic mechanisms can mediate the gene-environment dialog in early life and give rise to persistent epigenetic programming of adult physiology and dysfunction eventually resulting in disease. Understanding how early life experiences can give rise to lasting epigenetic marks conferring increased risk for mental disorders, how they are maintained and how they could be reversed, is increasingly becoming a focus of modern psychiatry and should pave new guidelines for timely therapeutic interventions.
Collapse
|
47
|
Babygirija R, Zheng J, Bülbül M, Cerjak D, Ludwig K, Takahashi T. Sustained delayed gastric emptying during repeated restraint stress in oxytocin knockout mice. J Neuroendocrinol 2010; 22:1181-6. [PMID: 20969650 DOI: 10.1111/j.1365-2826.2010.02069.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We have recently shown that impaired gastric motility observed in acute restraint stress was restored following repeated restraint stress in mice. Repeated restraint stress up-regulates oxytocin mRNA expression and down-regulates corticotrophin-releasing factor (CRF) mRNA expression at the hypothalamus. Oxytocin knockout mice (OXT-KO) have been widely used to study the central oxytocin signalling pathways in response to various stressors. We studied the effects of acute and repeated restraint stress on solid gastric emptying and hypothalamic CRF mRNA expression in wild-type (WT) and OXT-KO mice. Heterozygous (HZ) parents (B6; 129S-Oxt(tm1Wsy)/J mice) were bred in our animal facility. Male OXT-KO, WT and HZ littermates were used for the study. Solid gastric emptying was measured following acute restraint stress (for 90 min) or repeated restraint stress (for five consecutive days). Expression of CRF mRNA in the paraventricular nucleus (PVN) was measured by real-time reverse transcriptase-polymerase chain reaction. There were no significant differences of gastric emptying in WT (68.4 ± 4.1%, n = 6), HZ (71.8 ± 3.1%, n = 6) and OXT-KO (70.6 ± 3.1%, n = 6) mice in nonstressed conditions. Acute stress significantly delayed gastric emptying in OXT-KO mice (33.10 ± 2.5%, n = 6) WT (39.1 ± 1.1%, n = 6) and HZ mice (35.8 ± 1.2%, n = 6). Following repeated restraint stress loading, gastric emptying was significantly restored in WT (68.3 ± 4.5%, n = 6) and HZ mice (63.1 ± 2.6%, n = 6). By contrast, gastric emptying was still delayed in OXT-KO mice (34.7 ± 1.3%, n = 6) following repeated restraint stress. The increase in CRF mRNA expression at the PVN was much pronounced in OXT-KO mice compared to WT or HZ mice following repeated restraint stress. These findings suggest that central oxytocin plays a pivotal role in mediating the adaptation mechanism following repeated restraint stress in mice.
Collapse
Affiliation(s)
- R Babygirija
- Department of Surgery, Medical College of Wisconsin and Zablocki VA Medical Center, Milwaukee, WI 53295, USA
| | | | | | | | | | | |
Collapse
|
48
|
Neumann ID, Veenema AH, Beiderbeck DI. Aggression and anxiety: social context and neurobiological links. Front Behav Neurosci 2010; 4:12. [PMID: 20407578 PMCID: PMC2854527 DOI: 10.3389/fnbeh.2010.00012] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2009] [Accepted: 03/07/2010] [Indexed: 01/29/2023] Open
Abstract
Psychopathologies such as anxiety- and depression-related disorders are often characterized by impaired social behaviours including excessive aggression and violence. Excessive aggression and violence likely develop as a consequence of generally disturbed emotional regulation, such as abnormally high or low levels of anxiety. This suggests an overlap between brain circuitries and neurochemical systems regulating aggression and anxiety. In this review, we will discuss different forms of male aggression, rodent models of excessive aggression, and neurobiological mechanisms underlying male aggression in the context of anxiety. We will summarize our attempts to establish an animal model of high and abnormal aggression using rats selected for high (HAB) vs. low (LAB) anxiety-related behaviour. Briefly, male LAB rats and, to a lesser extent, male HAB rats show high and abnormal forms of aggression compared with non-selected (NAB) rats, making them a suitable animal model for studying excessive aggression in the context of extremes in innate anxiety. In addition, we will discuss differences in the activity of the hypothalamic–pituitary–adrenal axis, brain arginine vasopressin, and the serotonin systems, among others, which contribute to the distinct behavioural phenotypes related to aggression and anxiety. Further investigation of the neurobiological systems in animals with distinct anxiety phenotypes might provide valuable information about the link between excessive aggression and disturbed emotional regulation, which is essential for understanding the social and emotional deficits that are characteristic of many human psychiatric disorders.
Collapse
Affiliation(s)
- Inga D Neumann
- Department of Behavioural and Molecular Neuroendocrinology, University of Regensburg Regensburg, Germany
| | | | | |
Collapse
|
49
|
Frank E, Kessler MS, Filiou MD, Zhang Y, Maccarrone G, Reckow S, Bunck M, Heumann H, Turck CW, Landgraf R, Hambsch B. Stable isotope metabolic labeling with a novel N-enriched bacteria diet for improved proteomic analyses of mouse models for psychopathologies. PLoS One 2009; 4:e7821. [PMID: 19915716 PMCID: PMC2773927 DOI: 10.1371/journal.pone.0007821] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Accepted: 10/22/2009] [Indexed: 01/22/2023] Open
Abstract
The identification of differentially regulated proteins in animal models of psychiatric diseases is essential for a comprehensive analysis of associated psychopathological processes. Mass spectrometry is the most relevant method for analyzing differences in protein expression of tissue and body fluid proteomes. However, standardization of sample handling and sample-to-sample variability are problematic. Stable isotope metabolic labeling of a proteome represents the gold standard for quantitative mass spectrometry analysis. The simultaneous processing of a mixture of labeled and unlabeled samples allows a sensitive and accurate comparative analysis between the respective proteomes. Here, we describe a cost-effective feeding protocol based on a newly developed (15)N bacteria diet based on Ralstonia eutropha protein, which was applied to a mouse model for trait anxiety. Tissue from (15)N-labeled vs. (14)N-unlabeled mice was examined by mass spectrometry and differences in the expression of glyoxalase-1 (GLO1) and histidine triad nucleotide binding protein 2 (Hint2) proteins were correlated with the animals' psychopathological behaviors for methodological validation and proof of concept, respectively. Additionally, phenotyping unraveled an antidepressant-like effect of the incorporation of the stable isotope (15)N into the proteome of highly anxious mice. This novel phenomenon is of considerable relevance to the metabolic labeling method and could provide an opportunity for the discovery of candidate proteins involved in depression-like behavior. The newly developed (15)N bacteria diet provides researchers a novel tool to discover disease-relevant protein expression differences in mouse models using quantitative mass spectrometry.
Collapse
Affiliation(s)
- Elisabeth Frank
- Max Planck Institute of Psychiatry, Munich, Germany
- Schizophrenia Research Institute, School of Health Sciences, University of Wollongong, Wollongong, Australia
| | - Melanie S. Kessler
- Max Planck Institute of Psychiatry, Munich, Germany
- CNS Research, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | | | | | | | | | - Mirjam Bunck
- Max Planck Institute of Psychiatry, Munich, Germany
- Affectis Pharmaceuticals AG, Martinsried, Germany
| | - Hermann Heumann
- Max Planck Institute of Biochemistry, Martinsried, Germany
- Silantes GmbH, Munich, Germany
| | | | | | - Boris Hambsch
- Max Planck Institute of Psychiatry, Munich, Germany
- * E-mail:
| |
Collapse
|
50
|
Babygirija R, Zheng J, Ludwig K, Takahashi T. Central oxytocin is involved in restoring impaired gastric motility following chronic repeated stress in mice. Am J Physiol Regul Integr Comp Physiol 2009; 298:R157-65. [PMID: 19889866 DOI: 10.1152/ajpregu.00328.2009] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Accumulation of continuous life stress (chronic stress) often causes gastric symptoms. The development of gastric symptoms may depend on how humans adapt to the stressful events in their daily lives. Although acute stress delays gastric emptying and alters upper gastrointestinal motility in rodents, the effects of chronic stress on gastric motility and its adaptation mechanism remains unclear. Central oxytocin has been shown to have antistress effects. We studied whether central oxytocin is involved in mediating the adaptation mechanism following chronic repeated stress. Mice were loaded with acute and chronic stress (repeated stress for five consecutive days), and solid gastric emptying and postprandial gastric motility were compared between acute and chronic repeated stress. Expression of oxytocin and CRF mRNA in the hypothalamus was studied following acute and chronic repeated stress. Delayed gastric emptying during acute stress (43.1 +/- 7.8%; n = 6, P < 0.05) was completely restored to normal levels (72.1 +/- 2.4%; n = 6) following chronic repeated stress. Impaired gastric motility induced by acute stress was also restored following chronic repeated stress. Intracerebroventricular injection of oxytocin (0.1 and 0.5 microg) restored the impaired gastric emptying and motility induced by acute stress. The restored gastric emptying and motility following chronic repeated stress were antagonized by intracerebroventricular injection of oxytocin antagonists. Oxytocin mRNA expression in the supraoptic nucleus (SON) and paraventricular nucleus (PVN) of the hypothalamus was significantly increased following chronic repeated stress. In contrast, increased CRF mRNA expression in the SON and PVN in response to acute stress was significantly reduced following chronic repeated stress. Our study suggests the novel finding that the upregulation of central oxytocin expression is involved in mediating the adaptation mechanism following chronic repeated stress in mice.
Collapse
Affiliation(s)
- Reji Babygirija
- Department of Surgery, Medical College of Wisconsin and Zablocki VA Medical Center, Milwaukee, Wisconsin, USA
| | | | | | | |
Collapse
|