1
|
Kaplan HS, Horvath PM, Rahman MM, Dulac C. The neurobiology of parenting and infant-evoked aggression. Physiol Rev 2025; 105:315-381. [PMID: 39146250 DOI: 10.1152/physrev.00036.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 07/19/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024] Open
Abstract
Parenting behavior comprises a variety of adult-infant and adult-adult interactions across multiple timescales. The state transition from nonparent to parent requires an extensive reorganization of individual priorities and physiology and is facilitated by combinatorial hormone action on specific cell types that are integrated throughout interconnected and brainwide neuronal circuits. In this review, we take a comprehensive approach to integrate historical and current literature on each of these topics across multiple species, with a focus on rodents. New and emerging molecular, circuit-based, and computational technologies have recently been used to address outstanding gaps in our current framework of knowledge on infant-directed behavior. This work is raising fundamental questions about the interplay between instinctive and learned components of parenting and the mutual regulation of affiliative versus agonistic infant-directed behaviors in health and disease. Whenever possible, we point to how these technologies have helped gain novel insights and opened new avenues of research into the neurobiology of parenting. We hope this review will serve as an introduction for those new to the field, a comprehensive resource for those already studying parenting, and a guidepost for designing future studies.
Collapse
Affiliation(s)
- Harris S Kaplan
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| | - Patricia M Horvath
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| | - Mohammed Mostafizur Rahman
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| | - Catherine Dulac
- Department of Molecular and Cellular Biology, Howard Hughes Medical Institute, Center for Brain Science, Harvard University, Cambridge, Massachusetts, United States
| |
Collapse
|
2
|
Tsuda MC, Akoh-Arrey T, Mercurio JC, Rucker A, Airey ML, Jacobs H, Lukasz D, Wang L, Cameron HA. Adult Neurogenesis and the Initiation of Social Aggression in Male Mice. Hippocampus 2024. [PMID: 39376052 DOI: 10.1002/hipo.23643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/18/2024] [Accepted: 09/24/2024] [Indexed: 10/09/2024]
Abstract
The hippocampus is important for social behavior and exhibits unusual structural plasticity in the form of continued production of new granule neurons throughout adulthood, but it is unclear how adult neurogenesis contributes to social interactions. In the present study, we suppressed neurogenesis using a pharmacogenetic mouse model and examined social investigation and aggression in adult male mice to investigate the role of hippocampal adult-born neurons in the expression of aggressive behavior. In simultaneous choice tests with stimulus mice placed in corrals, mice with complete suppression of adult neurogenesis in adulthood (TK mice) exhibited normal social investigation behaviors, indicating that new neurons are not required for social interest, social memory, or detection of and response to social olfactory signals. However, mice with suppressed neurogenesis displayed decreased offensive and defensive aggression in a resident-intruder paradigm, and less resistance in a social dominance test, relative to neurogenesis-intact controls, when paired with weight and strain-matched (CD-1) mice. During aggression tests, TK mice were frequently attacked by the CD-1 intruder mice, which never occurred with WTs, and normal CD-1 male mice investigated TK mice less than controls when corralled in the social investigation test. Importantly, TK mice showed normal aggression toward prey (crickets) and smaller, nonaggressive (olfactory bulbectomized) C57BL/6J intruders, suggesting that mice lacking adult neurogenesis do not avoid aggressive social interactions if they are much larger than their opponent and will clearly win. Taken together, our findings show that adult hippocampal neurogenesis plays an important role in the instigation of intermale aggression, possibly by weighting a cost-benefit analysis against confrontation in cases where the outcome of the fight is not clear.
Collapse
Affiliation(s)
- Mumeko C Tsuda
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Talia Akoh-Arrey
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Jeffrey C Mercurio
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Ariana Rucker
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Megan L Airey
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Hannah Jacobs
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Daria Lukasz
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Lijing Wang
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Heather A Cameron
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
3
|
Taniguchi M, Murata Y, Yamaguchi M, Kaba H. Activation of arginine vasopressin receptor 1a reduces inhibitory synaptic currents at reciprocal synapses in the mouse accessory olfactory bulb. Front Cell Neurosci 2024; 18:1466817. [PMID: 39386179 PMCID: PMC11462548 DOI: 10.3389/fncel.2024.1466817] [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: 07/23/2024] [Accepted: 09/10/2024] [Indexed: 10/12/2024] Open
Abstract
Central arginine vasopressin (AVP) facilitates social recognition and modulates many complex social behaviors in mammals that, in many cases, recognize each other based on olfactory and/or pheromonal signals. AVP neurons are present in the accessory olfactory bulb (AOB), which is the first relay in the vomeronasal system and has been demonstrated to be a critical site for mating-induced mate recognition (olfactory memory) in female mice. The transmission of information from the AOB to higher centers is controlled by the dendrodendritic recurrent inhibition, i.e., inhibitory postsynaptic currents (IPSCs) generated in mitral cells by recurrent dendrodendritic inhibitory inputs from granule cells. These reports suggest that AVP might play an important role in regulating dendrodendritic inhibition in the AOB. To test this hypothesis, we examined the effects of extracellularly applied AVP on synaptic responses measured from mitral and granule cells in slice preparations from 23--36-day-old Balb/c mice. To evoke dendrodendritic inhibition in a mitral cell, depolarizing voltages of -70 to 0 mV (10 ms duration) were applied to a mitral cell using a conventional whole-cell configuration. We found that AVP significantly reduced the IPSCs. The suppressive effects of AVP on the IPSCs was diminished by an antagonist for vasopressin receptor 1a (V1aR) (Manning compound), but not by an antagonist for vasopressin receptor 1b (SSR149415). An agonist for V1aRs [(Phe2)OVT] mimicked the action of AVP on IPSCs. Additionally, AVP significantly suppressed voltage-activated currents in granule cells without affecting the magnitude of the response of mitral cells to gamma-aminobutyric acid (GABA). The present results suggest that V1aRs play a role in reciprocal transmission between mitral cells and granule cells in the mouse AOB by reducing GABAergic transmission through a presynaptic mechanism in granule cells.
Collapse
Affiliation(s)
- Mutsuo Taniguchi
- Department of Physiology, Kochi Medical School, Kochi University, Nankoku, Japan
| | | | | | | |
Collapse
|
4
|
Villegas A, Siegelbaum SA. Modulation of aggression by social novelty recognition memory in the hippocampal CA2 region. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592403. [PMID: 38746353 PMCID: PMC11092780 DOI: 10.1101/2024.05.03.592403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The dorsal CA2 subregion (dCA2) of the hippocampus exerts a critical role in social novelty recognition (SNR) memory and in the promotion of social aggression. Whether the social aggression and SNR memory functions of dCA2 are related or represent independent processes is unknown. Here we investigated the hypotheses that an animal is more likely to attack a novel compared to familiar animal and that dCA2 promotes social aggression through its ability to discriminate between novel and familiar conspecifics. To test these ideas, we conducted a multi-day resident intruder (R-I) test of aggression towards novel and familiar conspecifics. We found that mice were more likely to attack a novel compared to familiarized intruder and that silencing of dCA2 caused a more profound inhibition of aggression towards a novel than familiarized intruder. To explore whether and how dCA2 pyramidal neurons encode aggression, we recorded their activity using microendoscopic calcium imaging throughout the days of the R-I test. We found that a fraction of dCA2 neurons were selectively activated or inhibited during exploration, dominance, and attack behaviors and that these signals were enhanced during interaction with a novel compared to familiarized conspecific. Based on dCA2 population activity, a set of binary linear classifiers accurately decoded whether an animal was engaged in each of these forms of social behavior. Of particular interest, the accuracy of decoding aggression was greater with novel compared to familiarized intruders, with significant cross-day decoding using the same familiar animal on each day but not for a familiar-novel pair. Together, these findings demonstrate that dCA2 integrates information about social novelty with signals of behavioral state to promote aggression towards novel conspecifics.
Collapse
|
5
|
Siegler PN, Shaughnessy EK, Horman B, Vierling TT, King DH, Patisaul HB, Huhman KL, Alexander GM, Dudek SM. Identification of hippocampal area CA2 in hamster and vole brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.12.579957. [PMID: 38405991 PMCID: PMC10888814 DOI: 10.1101/2024.02.12.579957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Prairie voles (Microtus ochrogaster) and Syrian, or golden, hamsters (Mesocricetus auratus) are closely related to mice (Mus musculus) and rats (Rattus norvegicus, for example) and are commonly used in studies of social behavior including social interaction, social memory, and aggression. The CA2 region of the hippocampus is known to play a key role in social memory and aggression in mice and responds to social stimuli in rats, likely owing to its high expression of oxytocin and vasopressin 1b receptors. However, CA2 has yet to be identified and characterized in hamsters or voles. In this study, we sought to determine whether CA2 could be identified molecularly in vole and hamster. To do this, we used immunofluorescence with primary antibodies raised against known molecular markers of CA2 in mice and rats to stain hippocampal sections from voles and hamsters in parallel with those from mice. Here, we report that, like in mouse and rat, staining for many CA2 proteins in vole and hamster hippocampus reveals a population of neurons that express regulator of G protein signaling 14 (RGS14), Purkinje cell protein 4 (PCP4) and striatal-enriched protein tyrosine phosphatase (STEP), which together delineate the borders with CA3 and CA1. These cells were located at the distal end of the mossy fiber projections, marked by the presence of Zinc Transporter 3 (ZnT-3) and calbindin in all three species. In addition to staining the mossy fibers, calbindin also labeled a layer of CA1 pyramidal cells in mouse and hamster but not in vole. However, Wolframin ER transmembrane glycoprotein (WFS1) immunofluorescence, which marks all CA1 neurons, was present in all three species and abutted the distal end of CA2, marked by RGS14 immunofluorescence. Staining for two stress hormone receptors-the glucocorticoid (GR) and mineralocorticoid (MR) receptors-was also similar in all three species, with GR staining found primarily in CA1 and MR staining enriched in CA2. Interestingly, although perineuronal nets (PNNs) are known to surround CA2 cells in mouse and rat, we found that staining for PNNs differed across species in that both CA2 and CA3 showed staining in voles and primarily CA3 in hamsters with only some neurons in proximal CA2 showing staining. These results demonstrate that, like in mouse, CA2 in voles and hamsters can be molecularly distinguished from neighboring CA1 and CA3 areas, but PNN staining is less useful for identifying CA2 in the latter two species. These findings reveal commonalities across species in molecular profile of CA2, which will facilitate future studies of CA2 in these species. Yet to be determined is how differences in PNNs might relate to differences in social behavior across species.
Collapse
Affiliation(s)
- Preston Nicole Siegler
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709 USA
- Neuroscience Center, University of North Carolina, Chapel Hill, NC
| | | | - Brian Horman
- Department of Biological Sciences, North Carolina State University, Raleigh, NC
| | - Tia T. Vierling
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709 USA
| | - Darron H. King
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709 USA
| | - Heather B. Patisaul
- Department of Biological Sciences, North Carolina State University, Raleigh, NC
| | - Kim L. Huhman
- Neuroscience Institute, Georgia State University, Atlanta, GA
| | - Georgia M. Alexander
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709 USA
| | - Serena M. Dudek
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709 USA
| |
Collapse
|
6
|
Ritz NL, Brocka M, Butler MI, Cowan CSM, Barrera-Bugueño C, Turkington CJR, Draper LA, Bastiaanssen TFS, Turpin V, Morales L, Campos D, Gheorghe CE, Ratsika A, Sharma V, Golubeva AV, Aburto MR, Shkoporov AN, Moloney GM, Hill C, Clarke G, Slattery DA, Dinan TG, Cryan JF. Social anxiety disorder-associated gut microbiota increases social fear. Proc Natl Acad Sci U S A 2024; 121:e2308706120. [PMID: 38147649 PMCID: PMC10769841 DOI: 10.1073/pnas.2308706120] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 10/05/2023] [Indexed: 12/28/2023] Open
Abstract
Social anxiety disorder (SAD) is a crippling psychiatric disorder characterized by intense fear or anxiety in social situations and their avoidance. However, the underlying biology of SAD is unclear and better treatments are needed. Recently, the gut microbiota has emerged as a key regulator of both brain and behaviour, especially those related to social function. Moreover, increasing data supports a role for immune function and oxytocin signalling in social responses. To investigate whether the gut microbiota plays a causal role in modulating behaviours relevant to SAD, we transplanted the microbiota from SAD patients, which was identified by 16S rRNA sequencing to be of a differential composition compared to healthy controls, to mice. Although the mice that received the SAD microbiota had normal behaviours across a battery of tests designed to assess depression and general anxiety-like behaviours, they had a specific heightened sensitivity to social fear, a model of SAD. This distinct heightened social fear response was coupled with changes in central and peripheral immune function and oxytocin expression in the bed nucleus of the stria terminalis. This work demonstrates an interkingdom basis for social fear responses and posits the microbiome as a potential therapeutic target for SAD.
Collapse
Affiliation(s)
- Nathaniel L. Ritz
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Anatomy and Neuroscience, University College Cork, CorkT12YT20, Ireland
| | - Marta Brocka
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
| | - Mary I. Butler
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, CorkT12YT20, Ireland
| | - Caitlin S. M. Cowan
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
| | - Camila Barrera-Bugueño
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
| | - Christopher J. R. Turkington
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- School of Microbiology, University College Cork, CorkT12K8AF, Ireland
| | - Lorraine A. Draper
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- School of Microbiology, University College Cork, CorkT12K8AF, Ireland
| | - Thomaz F. S. Bastiaanssen
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Anatomy and Neuroscience, University College Cork, CorkT12YT20, Ireland
| | - Valentine Turpin
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
| | - Lorena Morales
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
| | - David Campos
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
| | - Cassandra E. Gheorghe
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Anatomy and Neuroscience, University College Cork, CorkT12YT20, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, CorkT12YT20, Ireland
| | - Anna Ratsika
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Anatomy and Neuroscience, University College Cork, CorkT12YT20, Ireland
| | - Virat Sharma
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- School of Microbiology, University College Cork, CorkT12K8AF, Ireland
| | - Anna V. Golubeva
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
| | - Maria R. Aburto
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Anatomy and Neuroscience, University College Cork, CorkT12YT20, Ireland
| | - Andrey N. Shkoporov
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- School of Microbiology, University College Cork, CorkT12K8AF, Ireland
| | - Gerard M. Moloney
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Anatomy and Neuroscience, University College Cork, CorkT12YT20, Ireland
| | - Colin Hill
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- School of Microbiology, University College Cork, CorkT12K8AF, Ireland
| | - Gerard Clarke
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, CorkT12YT20, Ireland
| | - David A. Slattery
- Department of Psychiatry, Psychosomatics and Psychotherapy, University Hospital Frankfurt, Frankfurt60528, Germany
| | - Timothy G. Dinan
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, CorkT12YT20, Ireland
| | - John F. Cryan
- Alimentary Pharmabiotic Centre Microbiome Ireland, University College Cork, CorkT12YT20, Ireland
- Department of Anatomy and Neuroscience, University College Cork, CorkT12YT20, Ireland
| |
Collapse
|
7
|
Wang Z, Yueh H, Chau M, Veenstra-VanderWeele J, O'Reilly KC. Circuits underlying social function and dysfunction. Autism Res 2023; 16:1268-1288. [PMID: 37458578 DOI: 10.1002/aur.2978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/13/2023] [Indexed: 08/01/2023]
Abstract
Substantial advances have been made toward understanding the genetic and environmental risk factors for autism, a neurodevelopmental disorder with social impairment as a core feature. In combination with optogenetic and chemogenetic tools to manipulate neural circuits in vivo, it is now possible to use model systems to test how specific neural circuits underlie social function and dysfunction. Here, we review the literature that has identified circuits associated with social interest (sociability), social reward, social memory, dominance, and aggression, and we outline a preliminary roadmap of the neural circuits driving these social behaviors. We highlight the neural circuitry underlying each behavioral domain, as well as develop an interactive map of how these circuits overlap across domains. We find that some of the circuits underlying social behavior are general and are involved in the control of multiple behavioral aspects, whereas other circuits appear to be specialized for specific aspects of social behavior. Our overlapping circuit map therefore helps to delineate the circuits involved in the various domains of social behavior and to identify gaps in knowledge.
Collapse
Affiliation(s)
- Ziwen Wang
- Department of Psychiatry, Columbia University; New York State Psychiatric Institute, New York, New York, USA
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hannah Yueh
- Department of Psychiatry, Columbia University; New York State Psychiatric Institute, New York, New York, USA
| | - Mirabella Chau
- Department of Psychiatry, Columbia University; New York State Psychiatric Institute, New York, New York, USA
| | - Jeremy Veenstra-VanderWeele
- Department of Psychiatry, Columbia University; New York State Psychiatric Institute, New York, New York, USA
| | - Kally C O'Reilly
- Department of Psychiatry, Columbia University; New York State Psychiatric Institute, New York, New York, USA
| |
Collapse
|
8
|
Gorlova A, Svirin E, Pavlov D, Cespuglio R, Proshin A, Schroeter CA, Lesch KP, Strekalova T. Understanding the Role of Oxidative Stress, Neuroinflammation and Abnormal Myelination in Excessive Aggression Associated with Depression: Recent Input from Mechanistic Studies. Int J Mol Sci 2023; 24:915. [PMID: 36674429 PMCID: PMC9861430 DOI: 10.3390/ijms24020915] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/26/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Aggression and deficient cognitive control problems are widespread in psychiatric disorders, including major depressive disorder (MDD). These abnormalities are known to contribute significantly to the accompanying functional impairment and the global burden of disease. Progress in the development of targeted treatments of excessive aggression and accompanying symptoms has been limited, and there exists a major unmet need to develop more efficacious treatments for depressed patients. Due to the complex nature and the clinical heterogeneity of MDD and the lack of precise knowledge regarding its pathophysiology, effective management is challenging. Nonetheless, the aetiology and pathophysiology of MDD has been the subject of extensive research and there is a vast body of the latest literature that points to new mechanisms for this disorder. Here, we overview the key mechanisms, which include neuroinflammation, oxidative stress, insulin receptor signalling and abnormal myelination. We discuss the hypotheses that have been proposed to unify these processes, as many of these pathways are integrated for the neurobiology of MDD. We also describe the current translational approaches in modelling depression, including the recent advances in stress models of MDD, and emerging novel therapies, including novel approaches to management of excessive aggression, such as anti-diabetic drugs, antioxidant treatment and herbal compositions.
Collapse
Affiliation(s)
- Anna Gorlova
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Laboratory of Cognitive Dysfunctions, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
| | - Evgeniy Svirin
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Laboratory of Cognitive Dysfunctions, Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
- Neuroplast BV, 6222 NK Maastricht, The Netherlands
| | - Dmitrii Pavlov
- Hotchkiss Brain Institute, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Raymond Cespuglio
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Centre de Recherche en Neurosciences de Lyon (CRNL), 69500 Bron, France
| | - Andrey Proshin
- P.K. Anokhin Research Institute of Normal Physiology, 125315 Moscow, Russia
| | - Careen A. Schroeter
- Preventive and Environmental Medicine, Kastanienhof Clinic, 50858 Köln-Junkersdorf, Germany
| | - Klaus-Peter Lesch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, The Netherlands
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Tatyana Strekalova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Maastricht University, 6229 ER Maastricht, The Netherlands
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
9
|
Svirin E, Veniaminova E, Costa-Nunes JP, Gorlova A, Umriukhin A, Kalueff AV, Proshin A, Anthony DC, Nedorubov A, Tse ACK, Walitza S, Lim LW, Lesch KP, Strekalova T. Predation Stress Causes Excessive Aggression in Female Mice with Partial Genetic Inactivation of Tryptophan Hydroxylase-2: Evidence for Altered Myelination-Related Processes. Cells 2022; 11:1036. [PMID: 35326487 PMCID: PMC8947002 DOI: 10.3390/cells11061036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 01/27/2023] Open
Abstract
The interaction between brain serotonin (5-HT) deficiency and environmental adversity may predispose females to excessive aggression. Specifically, complete inactivation of the gene encoding tryptophan hydroxylase-2 (Tph2) results in the absence of neuronal 5-HT synthesis and excessive aggressiveness in both male and female null mutant (Tph2-/-) mice. In heterozygous male mice (Tph2+/-), there is a moderate reduction in brain 5-HT levels, and when they are exposed to stress, they exhibit increased aggression. Here, we exposed female Tph2+/- mice to a five-day rat predation stress paradigm and assessed their emotionality and social interaction/aggression-like behaviors. Tph2+/- females exhibited excessive aggression and increased dominant behavior. Stressed mutants displayed altered gene expression of the 5-HT receptors Htr1a and Htr2a, glycogen synthase kinase-3 β (GSK-3β), and c-fos as well as myelination-related transcripts in the prefrontal cortex: myelin basic protein (Mbp), proteolipid protein 1 (Plp1), myelin-associated glycoprotein (Mag), and myelin oligodendrocyte glycoprotein (Mog). The expression of the plasticity markers synaptophysin (Syp) and cAMP response element binding protein (Creb), but not AMPA receptor subunit A2 (GluA2), were affected by genotype. Moreover, in a separate experiment, naïve female Tph2+/- mice showed signs of enhanced stress resilience in the modified swim test with repeated swimming sessions. Taken together, the combination of a moderate reduction in brain 5-HT with environmental challenges results in behavioral changes in female mice that resemble the aggression-related behavior and resilience seen in stressed male mutants; additionally, the combination is comparable to the phenotype of null mutants lacking neuronal 5-HT. Changes in myelination-associated processes are suspected to underpin the molecular mechanisms leading to aggressive behavior.
Collapse
Affiliation(s)
- Evgeniy Svirin
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands; (E.S.); (K.-P.L.)
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, 97080 Würzburg, Germany
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
| | - Ekaterina Veniaminova
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov University, 119991 Moscow, Russia; (E.V.); (J.P.C.-N.); (A.G.); (A.U.); (D.C.A.)
| | - João Pedro Costa-Nunes
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov University, 119991 Moscow, Russia; (E.V.); (J.P.C.-N.); (A.G.); (A.U.); (D.C.A.)
- Institute of Molecular Medicine, New University of Lisbon, 1649-028 Lisbon, Portugal
| | - Anna Gorlova
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov University, 119991 Moscow, Russia; (E.V.); (J.P.C.-N.); (A.G.); (A.U.); (D.C.A.)
| | - Aleksei Umriukhin
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov University, 119991 Moscow, Russia; (E.V.); (J.P.C.-N.); (A.G.); (A.U.); (D.C.A.)
| | - Allan V. Kalueff
- Neuroscience Program, Sirius University, 354340 Sochi, Russia;
- Moscow Institute of Physics and Technology, School of Biological and Medical Physics, 141701 Dolgoprudny, Russia
- Institute of Natural Sciences, Ural Federal University, 620002 Yekaterinburg, Russia
| | - Andrey Proshin
- P.K. Anokhin Research Institute of Normal Physiology, 125315 Moscow, Russia;
| | - Daniel C. Anthony
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov University, 119991 Moscow, Russia; (E.V.); (J.P.C.-N.); (A.G.); (A.U.); (D.C.A.)
- Department of Pharmacology, Oxford University, Oxford OX1 3QT, UK
| | - Andrey Nedorubov
- Institute of Translational Medicine and Biotechnology, Sechenov University, 119991 Moscow, Russia;
| | - Anna Chung Kwan Tse
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China;
| | - Susanne Walitza
- Department for Child and Adolescent Psychiatry and Psychotherapy, University Hospital of Psychiatry Zurich, University of Zurich, 8032 Zurich, Switzerland;
| | - Lee Wei Lim
- Li Ka Shing Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China;
| | - Klaus-Peter Lesch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands; (E.S.); (K.-P.L.)
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, 97080 Würzburg, Germany
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov University, 119991 Moscow, Russia; (E.V.); (J.P.C.-N.); (A.G.); (A.U.); (D.C.A.)
| | - Tatyana Strekalova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6200 MD Maastricht, The Netherlands; (E.S.); (K.-P.L.)
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, 125315 Moscow, Russia
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov University, 119991 Moscow, Russia; (E.V.); (J.P.C.-N.); (A.G.); (A.U.); (D.C.A.)
| |
Collapse
|
10
|
Cid-Jofré V, Moreno M, Reyes-Parada M, Renard GM. Role of Oxytocin and Vasopressin in Neuropsychiatric Disorders: Therapeutic Potential of Agonists and Antagonists. Int J Mol Sci 2021; 22:ijms222112077. [PMID: 34769501 PMCID: PMC8584779 DOI: 10.3390/ijms222112077] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/20/2021] [Accepted: 11/02/2021] [Indexed: 12/27/2022] Open
Abstract
Oxytocin (OT) and vasopressin (AVP) are hypothalamic neuropeptides classically associated with their regulatory role in reproduction, water homeostasis, and social behaviors. Interestingly, this role has expanded in recent years and has positioned these neuropeptides as therapeutic targets for various neuropsychiatric diseases such as autism, addiction, schizophrenia, depression, and anxiety disorders. Due to the chemical-physical characteristics of these neuropeptides including short half-life, poor blood-brain barrier penetration, promiscuity for AVP and OT receptors (AVP-R, OT-R), novel ligands have been developed in recent decades. This review summarizes the role of OT and AVP in neuropsychiatric conditions, as well as the findings of different OT-R and AVP-R agonists and antagonists, used both at the preclinical and clinical level. Furthermore, we discuss their possible therapeutic potential for central nervous system (CNS) disorders.
Collapse
Affiliation(s)
- Valeska Cid-Jofré
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile (USACH), Santiago 9170022, Chile; (V.C.-J.); (M.M.)
| | - Macarena Moreno
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile (USACH), Santiago 9170022, Chile; (V.C.-J.); (M.M.)
- Facultad de Ciencias Sociales, Escuela de Psicología, Universidad Bernardo OHiggins, Santiago 8370993, Chile
| | - Miguel Reyes-Parada
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile (USACH), Santiago 9170022, Chile; (V.C.-J.); (M.M.)
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Providencia 7500912, Chile
- Correspondence: (M.R.-P.); (G.M.R.)
| | - Georgina M. Renard
- Centro de Investigación Biomédica y Aplicada (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile (USACH), Santiago 9170022, Chile; (V.C.-J.); (M.M.)
- Correspondence: (M.R.-P.); (G.M.R.)
| |
Collapse
|
11
|
Aulino EA, Caldwell HK. Pharmacological manipulation of oxytocin receptor signaling during mouse embryonic development results in sex-specific behavioral effects in adulthood. Horm Behav 2021; 135:105026. [PMID: 34273706 PMCID: PMC8487944 DOI: 10.1016/j.yhbeh.2021.105026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 12/01/2022]
Abstract
The oxytocin (Oxt) system is a known neuromodulator of social behaviors, but also appears to contribute to the development of sex-specific neural circuitry. In this latter role, the Oxt system helps to lay the foundation for sex-specific behaviors across the life span. In mice, the Oxt system emerges in early development, with sex differences in the expression of Oxt and a temporal offset in the expression of the Oxt receptor (Oxtr) relative to Oxt. In females, Oxt mRNA is detectable by embryonic day (E) 16.5, but in males, Oxt mRNA is not measurable until after birth. However, in both sexes, Oxtr mRNA is detectable by E12.5 and binding by E16.5. While the postnatal Oxt system has been studied, little is known about the embryonic Oxt system. Therefore, we hypothesize that it directly contributes to the developmental trajectory of the brain, ultimately affecting adult sex-specific behaviors. To test this hypothesis, Oxtr signaling was transiently disrupted at E16.5 using an Oxtr antagonist (OxtrA) and the effects on adult behavior evaluated. OxtrA-treated adult males displayed increased agonistic behavior, social investigation, and depressive-like behavior compared to vehicle-injected controls, while OxtrA-treated adult females had impaired social recognition memory compared to vehicle-injected controls. These data are the first to identify a functional link between the organizational activity of the embryonic Oxt system and adult behavior. Further, this work suggests that the Oxt system does more than serve as a neuromodulator in adulthood, but rather, may help shape the development of the neural circuitry regulating sex-specific behaviors.
Collapse
Affiliation(s)
- Elizabeth A Aulino
- Laboratory of Neuroendocrinology and Behavior, Department of Biological Sciences, School of Biomedical Sciences, Brain Health Research Institute, Kent State University, Kent, OH, USA
| | - Heather K Caldwell
- Laboratory of Neuroendocrinology and Behavior, Department of Biological Sciences, School of Biomedical Sciences, Brain Health Research Institute, Kent State University, Kent, OH, USA.
| |
Collapse
|
12
|
Lv J, Hua SJ, Wu XF, Ding YJ, Zhang CL, Sun FJ. REMOVED: Antidiuretic hormone associates with the cognitive memory in small-cell lung cancer patients. CURRENT RESEARCH IN BEHAVIORAL SCIENCES 2021. [DOI: 10.1016/j.crbeha.2021.100061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
13
|
Lehr AB, Kumar A, Tetzlaff C, Hafting T, Fyhn M, Stöber TM. CA2 beyond social memory: Evidence for a fundamental role in hippocampal information processing. Neurosci Biobehav Rev 2021; 126:398-412. [PMID: 33775693 DOI: 10.1016/j.neubiorev.2021.03.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 01/16/2023]
Abstract
Hippocampal region CA2 has received increased attention due to its importance in social recognition memory. While its specific function remains to be identified, there are indications that CA2 plays a major role in a variety of situations, widely extending beyond social memory. In this targeted review, we highlight lines of research which have begun to converge on a more fundamental role for CA2 in hippocampus-dependent memory processing. We discuss recent proposals that speak to the computations CA2 may perform within the hippocampal circuit.
Collapse
Affiliation(s)
- Andrew B Lehr
- Department of Computational Neuroscience, University of Göttingen, Germany; Bernstein Center for Computational Neuroscience, University of Göttingen, Germany; Department of Computational Physiology, Simula Research Laboratory, Lysaker, Norway; Centre for Integrative Neuroplasticity, University of Oslo, Norway.
| | - Arvind Kumar
- Department of Computational Science and Technology, KTH Royal Institute of Technology, Sweden
| | - Christian Tetzlaff
- Department of Computational Neuroscience, University of Göttingen, Germany; Bernstein Center for Computational Neuroscience, University of Göttingen, Germany
| | - Torkel Hafting
- Centre for Integrative Neuroplasticity, University of Oslo, Norway; Institute of Basic Medical Sciences, University of Oslo, Norway
| | - Marianne Fyhn
- Centre for Integrative Neuroplasticity, University of Oslo, Norway; Department of Biosciences, University of Oslo, Norway
| | - Tristan M Stöber
- Department of Computational Physiology, Simula Research Laboratory, Lysaker, Norway; Centre for Integrative Neuroplasticity, University of Oslo, Norway; Department of Informatics, University of Oslo, Norway.
| |
Collapse
|
14
|
Sparapani S, Millet-Boureima C, Oliver J, Mu K, Hadavi P, Kalostian T, Ali N, Avelar CM, Bardies M, Barrow B, Benedikt M, Biancardi G, Bindra R, Bui L, Chihab Z, Cossitt A, Costa J, Daigneault T, Dault J, Davidson I, Dias J, Dufour E, El-Khoury S, Farhangdoost N, Forget A, Fox A, Gebrael M, Gentile MC, Geraci O, Gnanapragasam A, Gomah E, Haber E, Hamel C, Iyanker T, Kalantzis C, Kamali S, Kassardjian E, Kontos HK, Le TBU, LoScerbo D, Low YF, Mac Rae D, Maurer F, Mazhar S, Nguyen A, Nguyen-Duong K, Osborne-Laroche C, Park HW, Parolin E, Paul-Cole K, Peer LS, Philippon M, Plaisir CA, Porras Marroquin J, Prasad S, Ramsarun R, Razzaq S, Rhainds S, Robin D, Scartozzi R, Singh D, Fard SS, Soroko M, Soroori Motlagh N, Stern K, Toro L, Toure MW, Tran-Huynh S, Trépanier-Chicoine S, Waddingham C, Weekes AJ, Wisniewski A, Gamberi C. The Biology of Vasopressin. Biomedicines 2021; 9:89. [PMID: 33477721 PMCID: PMC7832310 DOI: 10.3390/biomedicines9010089] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/29/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
Vasopressins are evolutionarily conserved peptide hormones. Mammalian vasopressin functions systemically as an antidiuretic and regulator of blood and cardiac flow essential for adapting to terrestrial environments. Moreover, vasopressin acts centrally as a neurohormone involved in social and parental behavior and stress response. Vasopressin synthesis in several cell types, storage in intracellular vesicles, and release in response to physiological stimuli are highly regulated and mediated by three distinct G protein coupled receptors. Other receptors may bind or cross-bind vasopressin. Vasopressin is regulated spatially and temporally through transcriptional and post-transcriptional mechanisms, sex, tissue, and cell-specific receptor expression. Anomalies of vasopressin signaling have been observed in polycystic kidney disease, chronic heart failure, and neuropsychiatric conditions. Growing knowledge of the central biological roles of vasopressin has enabled pharmacological advances to treat these conditions by targeting defective systemic or central pathways utilizing specific agonists and antagonists.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Chiara Gamberi
- Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada; (S.S.); (C.M.-B.); (J.O.); (K.M.); (P.H.); (T.K.); (N.A.); (C.M.A.); (M.B.); (B.B.); (M.B.); (G.B.); (R.B.); (L.B.); (Z.C.); (A.C.); (J.C.); (T.D.); (J.D.); (I.D.); (J.D.); (E.D.); (S.E.-K.); (N.F.); (A.F.); (A.F.); (M.G.); (M.C.G.); (O.G.); (A.G.); (E.G.); (E.H.); (C.H.); (T.I.); (C.K.); (S.K.); (E.K.); (H.K.K.); (T.B.U.L.); (D.L.); (Y.F.L.); (D.M.R.); (F.M.); (S.M.); (A.N.); (K.N.-D.); (C.O.-L.); (H.W.P.); (E.P.); (K.P.-C.); (L.S.P.); (M.P.); (C.-A.P.); (J.P.M.); (S.P.); (R.R.); (S.R.); (S.R.); (D.R.); (R.S.); (D.S.); (S.S.F.); (M.S.); (N.S.M.); (K.S.); (L.T.); (M.W.T.); (S.T.-H.); (S.T.-C.); (C.W.); (A.J.W.); (A.W.)
| |
Collapse
|
15
|
Stöber TM, Lehr AB, Hafting T, Kumar A, Fyhn M. Selective neuromodulation and mutual inhibition within the
CA3–CA2
system can prioritize sequences for replay. Hippocampus 2020; 30:1228-1238. [DOI: 10.1002/hipo.23256] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/10/2020] [Accepted: 08/07/2020] [Indexed: 11/06/2022]
Affiliation(s)
- Tristan M. Stöber
- Department of Computational Physiology Simula Research Laboratory Lysaker Norway
- Centre for Integrative Neuroplasticity University of Oslo Oslo Norway
- Department of Informatics University of Oslo Oslo Norway
| | - Andrew B. Lehr
- Department of Computational Physiology Simula Research Laboratory Lysaker Norway
- Centre for Integrative Neuroplasticity University of Oslo Oslo Norway
- Department of Computational Neuroscience University of Göttingen Göttingen Germany
| | - Torkel Hafting
- Centre for Integrative Neuroplasticity University of Oslo Oslo Norway
- Institute of Basic Medical Sciences University of Oslo Oslo Norway
| | - Arvind Kumar
- Department of Computational Science and Technology KTH Royal Institute of Technology Stockholm Sweden
| | - Marianne Fyhn
- Centre for Integrative Neuroplasticity University of Oslo Oslo Norway
- Department of Biosciences University of Oslo Oslo Norway
| |
Collapse
|
16
|
Grattan DR, Ladyman SR. Neurophysiological and cognitive changes in pregnancy. HANDBOOK OF CLINICAL NEUROLOGY 2020; 171:25-55. [PMID: 32736755 DOI: 10.1016/b978-0-444-64239-4.00002-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The hormonal fluctuations in pregnancy drive a wide range of adaptive changes in the maternal brain. These range from specific neurophysiological changes in the patterns of activity of individual neuronal populations, through to complete modification of circuit characteristics leading to fundamental changes in behavior. From a neurologic perspective, the key hormone changes are those of the sex steroids, estradiol and progesterone, secreted first from the ovary and then from the placenta, the adrenal glucocorticoid cortisol, as well as the anterior pituitary peptide hormone prolactin and its pregnancy-specific homolog placental lactogen. All of these hormones are markedly elevated during pregnancy and cross the blood-brain barrier to exert actions on neuronal populations through receptors expressed in specific regions. Many of the hormone-induced changes are in autonomic or homeostatic systems. For example, patterns of oxytocin and prolactin secretion are dramatically altered to support novel physiological functions. Appetite is increased and feedback responses to metabolic hormones such as leptin and insulin are suppressed to promote a positive energy balance. Fundamental physiological systems such as glucose homeostasis and thermoregulation are modified to optimize conditions for fetal development. In addition to these largely autonomic changes, there are also changes in mood, behavior, and higher processes such as cognition. This chapter summarizes the hormonal changes associated with pregnancy and reviews how these changes impact on brain function, drawing on examples from animal research, as well as available information about human pregnancy.
Collapse
Affiliation(s)
- David R Grattan
- Centre for Neuroendocrinology and Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.
| | - Sharon R Ladyman
- Centre for Neuroendocrinology and Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
17
|
Williams Avram SK, Lee HJ, Fastman J, Cymerblit-Sabba A, Smith A, Vincent M, Song J, Granovetter MC, Lee SH, Cilz NI, Stackmann M, Chaturvedi R, Young WS. NMDA Receptor in Vasopressin 1b Neurons Is Not Required for Short-Term Social Memory, Object Memory or Aggression. Front Behav Neurosci 2019; 13:218. [PMID: 31787886 PMCID: PMC6856057 DOI: 10.3389/fnbeh.2019.00218] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/05/2019] [Indexed: 12/20/2022] Open
Abstract
The arginine vasopressin 1b receptor (Avpr1b) plays an important role in social behaviors including aggression, social learning and memory. Genetic removal of Avpr1b from mouse models results in deficits in aggression and short-term social recognition in adults. Avpr1b gene expression is highly enriched in the pyramidal neurons of the hippocampal cornu ammonis 2 (CA2) region. Activity of the hippocampal CA2 has been shown to be required for normal short-term social recognition and aggressive behaviors. Vasopressin acts to enhance synaptic responses of CA2 neurons through a NMDA-receptor dependent mechanism. Genetic removal of the obligatory subunit of the NMDA receptor (Grin1) within distinct hippocampal regions impairs non-social learning and memory. However, the question of a direct role for NMDA receptor activity in Avpr1b neurons to modulate social behavior remains unclear. To answer this question, we first created a novel transgenic mouse line with Cre recombinase knocked into the Avpr1b coding region to genetically target Avpr1b neurons. We confirmed this line has dense Cre expression throughout the dorsal and ventral CA2 regions of the hippocampus, along with scattered expression within the caudate-putamen and olfactory bulb (OB). Conditional removal of the NMDA receptor was achieved by crossing our line to an available floxed Grin1 line. The resulting mice were measured on a battery of social and memory behavioral tests. Surprisingly, we did not observe any differences between Avpr1b-Grin1 knockout mice and their wildtype siblings. We conclude that mice without typical NMDA receptor function in Avpr1b neurons can develop normal aggression as well as short-term social and object memory performance.
Collapse
Affiliation(s)
- Sarah K Williams Avram
- Section on Neural Gene Expression, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States.,Systems Neuroscience Imaging Resource, Intramural Research Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Heon-Jin Lee
- Section on Neural Gene Expression, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States.,Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Jarrett Fastman
- Section on Neural Gene Expression, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Adi Cymerblit-Sabba
- Section on Neural Gene Expression, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Adam Smith
- Section on Neural Gene Expression, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States.,Neuroscience Program, Department of Pharmacology & Toxicology, University of Kansas, Lawrence, KS, United States
| | - Matthew Vincent
- Section on Neural Gene Expression, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - June Song
- Section on Neural Gene Expression, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Michael C Granovetter
- Section on Neural Gene Expression, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Su-Hyun Lee
- Section on Neural Gene Expression, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Nicholas I Cilz
- Section on Neural Gene Expression, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Michelle Stackmann
- Section on Neural Gene Expression, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - Rahul Chaturvedi
- Section on Neural Gene Expression, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| | - W Scott Young
- Section on Neural Gene Expression, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
18
|
Kanitz E, Tuchscherer M, Otten W, Tuchscherer A, Zebunke M, Puppe B. Coping Style of Pigs Is Associated With Different Behavioral, Neurobiological and Immune Responses to Stressful Challenges. Front Behav Neurosci 2019; 13:173. [PMID: 31417378 PMCID: PMC6686684 DOI: 10.3389/fnbeh.2019.00173] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 07/15/2019] [Indexed: 12/14/2022] Open
Abstract
Based on the animal’s reaction to environmental challenges, consistent but different coping styles can be identified, which in turn may have consequences for health and welfare. Therefore, profound knowledge of the complex interrelationships between individual behavioral response patterns, underlying neurobiological mechanisms and immunological effects is required. The aim of this study was to examine whether pigs with different coping styles exhibit distinct behavioral, neurobiological and immune responses to stressful situations. Therefore, pigs (n = 40) were classified as proactive, reactive or intermediate animals according to a repeatedly-performed backtest, and behavioral, neuroendocrine and immune alterations were analyzed without any stress before weaning on day 28 and after a stress treatment on day 32. Our results show that the behavioral responses in an open-field/novel-object test characterized proactive pigs as more active. There were no significant differences in adrenocorticotropic hormone and cortisol concentrations between pigs with different coping characteristics. However, we found that proactive pigs displayed significantly increased plasma noradrenaline levels in response to stress, which may reflect a higher sympathetic reactivity of these animals. Furthermore, the present study revealed coping style differences in mRNA expression of mineralocorticoid, glucocorticoid, oxytocin and arginine vasopressin receptors and the immediate early gene c-fos in stress-related brain regions. While proactive pigs responded to stress with higher mRNA expression of arginine vasopressin, mineralocorticoid and glucocorticoid receptors, reactive pigs displayed higher oxytocin receptor and c-fos mRNA expression, indicating different neurobiological mechanisms of distinct coping styles in response to stressful challenges. Moreover, we also found humoral immune differences between proactive, intermediate and reactive animals. Proactive pigs had a higher total serum IgA concentration before and after stress treatment, with a significant increase in response to stress compared to reactive and intermediate pigs. In contrast, stress-induced IgM concentrations only increased in reactive and intermediate animals, suggesting that the effects of coping style on humoral immunity may differ depending on the specific function of the immunoglobulin classes. In conclusion, this multidisciplinary study expands the concept of coping style in farm animals, particularly in terms of individual stress reactivity and disease susceptibility, and thus contributes to the understanding of the biology of animal welfare.
Collapse
Affiliation(s)
- Ellen Kanitz
- Institute of Behavioural Physiology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Margret Tuchscherer
- Institute of Behavioural Physiology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Winfried Otten
- Institute of Behavioural Physiology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Armin Tuchscherer
- Institute of Genetics and Biometry, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Manuela Zebunke
- Institute of Behavioural Physiology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Birger Puppe
- Institute of Behavioural Physiology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany.,Behavioural Sciences, Faculty of Agricultural and Environmental Sciences, University of Rostock, Rostock, Germany
| |
Collapse
|
19
|
Smiley KO, Ladyman SR, Gustafson P, Grattan DR, Brown RSE. Neuroendocrinology and Adaptive Physiology of Maternal Care. Curr Top Behav Neurosci 2019; 43:161-210. [PMID: 31808002 DOI: 10.1007/7854_2019_122] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Parental care is critical for offspring survival in many species. In mammals, parental care is primarily provided through maternal care, due to obligate pregnancy and lactation constraints, although some species also show paternal and alloparental care. These behaviors are driven by specialized neural circuits that receive sensory, cortical, and hormonal input to generate a coordinated and timely change in behavior, and sustain that behavior through activation of reward pathways. Importantly, the hormonal changes associated with pregnancy and lactation also act to coordinate a broad range of physiological changes to support the mother and enable her to adapt to the demands of these states. This chapter will review the neural pathways that regulate maternal behavior, the hormonal changes that occur during pregnancy and lactation, and how these two facets merge together to promote both young-directed maternal responses (including nursing and grooming) and young-related responses (including maternal aggression and other physiological adaptions to support the development of and caring for young). We conclude by examining how experimental animal work has translated into knowledge of human parenting, particularly in regards to maternal mental health issues.
Collapse
Affiliation(s)
- Kristina O Smiley
- Centre for Neuroendocrinology and Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Sharon R Ladyman
- Centre for Neuroendocrinology and Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Papillon Gustafson
- Centre for Neuroendocrinology and Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - David R Grattan
- Centre for Neuroendocrinology and Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Rosemary S E Brown
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
20
|
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: 1.8] [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
|
21
|
Shimizu K, Nakamura K, Yokosuka M, Kondo Y. Modulation of male mouse sociosexual and anxiety-like behaviors by vasopressin receptors. Physiol Behav 2018; 197:37-41. [PMID: 30290180 DOI: 10.1016/j.physbeh.2018.09.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/18/2018] [Accepted: 09/28/2018] [Indexed: 11/30/2022]
Abstract
Although the involvement of two types of vasopressin (AVP) receptors, v1a and v1b, in neural regulation of social behavior is well documented in rodents, there is no report on combined actions of them in regulation of social behavior. In this study, we investigated behavioral differences between wild-type (WT) and v1a and v1b double knockout (dKO) mice. For this, we measured olfactory preference, sexual behavior with receptive females (four weekly tests) in an enriched large observation cage, and anxiety-like behaviors. No difference between WT and dKO mice was found in olfactory preferences for estrous female odor to male odor. Over all four mating tests, the number of mounts and pursuits after receptive females was significantly greater in dKO mice than in WT mice. In the elevated plus maze and the open field test, dKO mice showed lower anxiety-like behavior than WT mice. Finally, we measured approach behavior to several types of objects, figurines, and caged anestrous or estrous females placed in the open field apparatus. The only difference observed was that dKO mice spent longer in the vicinity of estrous females than did WT mice. These findings suggest that vasopressin receptors are involved in the regulation of sociosexual behavior, presumably partly mediated by emotional responses, in male mice.
Collapse
Affiliation(s)
- Kie Shimizu
- Department of Animal Sciences, Teikyo University of Science, Yamanashi, Japan; Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Kazuaki Nakamura
- Department of Pharmacology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Makoto Yokosuka
- Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Yasuhiko Kondo
- Department of Animal Sciences, Teikyo University of Science, Yamanashi, Japan.
| |
Collapse
|
22
|
Jager A, Maas DA, Fricke K, de Vries RB, Poelmans G, Glennon JC. Aggressive behavior in transgenic animal models: A systematic review. Neurosci Biobehav Rev 2018; 91:198-217. [DOI: 10.1016/j.neubiorev.2017.09.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 08/10/2017] [Accepted: 09/19/2017] [Indexed: 11/25/2022]
|
23
|
Fam BS, Paré P, Felkl AB, Vargas-Pinilla P, Paixão-Côrtes VR, Viscardi LH, Bortolini MC. Oxytocin and arginine vasopressin systems in the domestication process. Genet Mol Biol 2018; 41:235-242. [PMID: 29668014 PMCID: PMC5913714 DOI: 10.1590/1678-4685-gmb-2017-0069] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 10/01/2017] [Indexed: 11/22/2022] Open
Abstract
Domestication is of unquestionable importance to the technological revolution that has given rise to modern human societies. In this study, we analyzed the DNA and protein sequences of six genes of the oxytocin and arginine vasopressin systems (OXT-OXTR; AVP-AVPR1a, AVPR1b and AVPR2) in 40 placental mammals. These systems play an important role in the control of physiology and behavior. According to our analyses, neutrality does not explain the pattern of molecular evolution found in some of these genes. We observed specific sites under positive selection in AVPR1b (ω = 1.429, p = 0.001) and AVPR2 (ω= 1.49, p = 0.001), suggesting that they could be involved in behavior and physiological changes, including those related to the domestication process. Furthermore, AVPR1a, which plays a role in social behavior, is under relaxed selective constraint in domesticated species. These results provide new insights into the nature of the domestication process and its impact on the OXT-AVP system.
Collapse
Affiliation(s)
- Bibiana S.O. Fam
- Departamento de Genética, Universidade Federal do Rio Grande do
Sul, Porto Alegre, RS, Brazil
| | - Pamela Paré
- Departamento de Genética, Universidade Federal do Rio Grande do
Sul, Porto Alegre, RS, Brazil
| | - Aline B. Felkl
- Departamento de Genética, Universidade Federal do Rio Grande do
Sul, Porto Alegre, RS, Brazil
| | - Pedro Vargas-Pinilla
- Departamento de Genética, Universidade Federal do Rio Grande do
Sul, Porto Alegre, RS, Brazil
| | | | | | - Maria Cátira Bortolini
- Departamento de Genética, Universidade Federal do Rio Grande do
Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
24
|
Ivask M, Pajusalu S, Reimann E, Kõks S. Hippocampus and Hypothalamus RNA-sequencing of WFS1-deficient Mice. Neuroscience 2018; 374:91-103. [PMID: 29406269 DOI: 10.1016/j.neuroscience.2018.01.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 11/26/2017] [Accepted: 01/18/2018] [Indexed: 01/08/2023]
Abstract
Wolfram syndrome is caused by mutations in the WFS1 gene. WFS1 protein dysfunction results in a range of neuroendocrine syndromes and is mostly characterized by juvenile-onset diabetes mellitus and optic atrophy. WFS1 has been shown to participate in membrane trafficking, protein processing and Ca2+ homeostasis in the endoplasmic reticulum. Aim of the present study was to find the transcriptomic changes influenced by WFS1 in the hypothalamus and hippocampus using RNA-sequencing. The WFS1-deficient mice were used as a model system to analyze the changes in transcriptional networks. The number of differentially expressed genes between hypothalami of WFS1-deficient (Wfs1KO) and wild-type (WT) mice was 43 and between hippocampi 311 with False Discovery Rate (FDR) <0.05. Avpr1a and Avpr1b were significantly upregulated in the hypothalamus and hippocampus of Wfs1KO mice respectively. Trpm8 was the most upregulated gene in the hippocampus of Wfs1KO mice. The functional analysis revealed significant enrichment of networks and pathways associated with protein synthesis, cell-to-cell signaling and interaction, molecular transport, metabolic disease and nervous system development and function. In conclusion, the transcriptomic profiles of WFS1-deficient hypothalamus and hippocampus do indicate the activation of degenerative molecular pathways causing the clinical occurrences typical to Wolfram syndrome.
Collapse
Affiliation(s)
- Marilin Ivask
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia.
| | - Sander Pajusalu
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu, Estonia; Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Ene Reimann
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Sulev Kõks
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
25
|
Kutlubaev MA, Mendelevich VD. The problem of aggressive behavior in epilepsy: clinical and neurobiological aspects. Zh Nevrol Psikhiatr Im S S Korsakova 2018; 118:94-100. [DOI: 10.17116/jnevro20181187194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
26
|
Panagiotidis D, Clemens B, Habel U, Schneider F, Schneider I, Wagels L, Votinov M. Exogenous testosterone in a non-social provocation paradigm potentiates anger but not behavioral aggression. Eur Neuropsychopharmacol 2017; 27:1172-1184. [PMID: 28939164 DOI: 10.1016/j.euroneuro.2017.07.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 06/16/2017] [Accepted: 07/18/2017] [Indexed: 10/18/2022]
Abstract
Animal studies suggest a causal link between testosterone and aggression. However, in human research the exact role of this hormone is still unclear, having been linked to dominance and approach behavior rather than to aggression per se. In a social context, the induction of aggression might be confounded with dominance or status changes, which potentially influence the association between aggression and testosterone. The objective of the current study was to investigate the influence of testosterone on non-social aggression in a double-blind, placebo-controlled experiment including 90 healthy male participants. To this end, we developed an innovative paradigm in which participants were provoked by a malfunctioning joystick restraining them from a promised reward. As measures for aggression throughout the task the joystick amplitude was recorded and anger was assessed via emotional self-ratings. Participants reacted to the provocation with a significant shift to more negative emotions and increased implicit aggressive behavior, reflected in the force exerted to pull the joystick following provocation. Importantly, the study demonstrated first evidence for a modulating influence of testosterone on non-social aggression in males: Self-rated anger was significantly elevated in the testosterone group compared to the placebo group as a function of provocation. Testosterone administration did not significantly influence the implicit aggressive response. These findings demonstrate a potentiating effect of testosterone on provocation-related anger in a non-social context. Furthermore, the results highlight the importance of disentangling different components of aggression and characterizing different influencing factors when inferring on hormonal effects.
Collapse
Affiliation(s)
- Despina Panagiotidis
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | - Benjamin Clemens
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Ute Habel
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany; JARA-Institute Brain Structure-Function Relationship, Research Center Jülich and RWTH Aachen University, 52074 Aachen, Germany
| | - Frank Schneider
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany; JARA-Institute Brain Structure-Function Relationship, Research Center Jülich and RWTH Aachen University, 52074 Aachen, Germany
| | - Isabella Schneider
- Department of General Psychiatry, Center of Psychosocial Medicine, University of Heidelberg, Germany
| | - Lisa Wagels
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Mikhail Votinov
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, Uniklinik RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany; Institute of Neuroscience and Medicine 10, Research Center Jülich, Jülich, Germany
| |
Collapse
|
27
|
Geng CH, Wang C, Yang J, Wang H, Ma RQ, Liu X, Wang CH. Arginine vasopressin improves the memory deficits in Han Chinese patients with first-episode schizophrenia. Peptides 2017; 97:8-15. [PMID: 28882471 DOI: 10.1016/j.peptides.2017.09.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 08/29/2017] [Accepted: 09/02/2017] [Indexed: 11/18/2022]
Abstract
The memory impairment is a core deficit in the first-episode schizophrenia patients. Arginine vasopressin (AVP) in the brain can improve learning and memory. We performed multicentre, randomized, double-blind, placebo-controlled, parallel-group clinical trial to study the cognitive functioning in Han Chinese first-episode schizophrenic patients in a 12-week treatment regime with the intranasal administration of AVP (128 cases) or placebo (131 cases) in addition to the conventional treatment. The methods of positive and negative syndrome scale (PANSS), Wechsler memory scale-4th edition (WMS-IV) and event-related potential (ERP) were used to study the effects of AVP on the cognitive function. The results showed that (1) AVP concentration decreased in cerebrospinal fluid (CSF) of the right-handed Han Chinese first-episode schizophrenic patients comparing with that of the health volunteers (7.1±1.5pg/ml vs 13.3±1.9pg/ml, p<0.01), and did not change in plasma; (2) AVP significantly improved PANSS scores including total scores, positive symptoms, negative symptoms and general psychopathology comparing with those of the placebo group; (3) AVP elevated WMS-IV scores including the long-term memory (accumulation), short-term memory (recognition, comprehension), immediate memory (number recitation) and memory quotient 4, 8 and 12 weeks after treatment; and (4) AVP did not influence the latency and wave amplitude of target stimulus of P300 of right-handed Han Chinese first-episode schizophrenic patients. The data suggested that AVP might improve cognitive process, such as memorizing and extraction of the information although there were many changes of cognitive functions in the right-handed Han Chinese first-episode schizophrenic patients.
Collapse
Affiliation(s)
- Cai-Hong Geng
- The Second Affiliated Hospital of Xinxiang Medical University and Henan Province Mental Hospital, Xinxiang, Henan 453002, China
| | - Chao Wang
- The Second Affiliated Hospital of Xinxiang Medical University and Henan Province Mental Hospital, Xinxiang, Henan 453002, China
| | - Jun Yang
- Xinxiang Institute for New Medicine, Xinxing, Henan 453003, China; Xinjiang Hongda Food & Beverage Ltd., Xinjiang, Shanxi 043110, China.
| | - Hua Wang
- Xinxiang Institute for New Medicine, Xinxing, Henan 453003, China; Xinjiang Hongda Food & Beverage Ltd., Xinjiang, Shanxi 043110, China
| | - Rui-Qing Ma
- Xinxiang Institute for New Medicine, Xinxing, Henan 453003, China
| | - Xu Liu
- The Second Affiliated Hospital of Xinxiang Medical University and Henan Province Mental Hospital, Xinxiang, Henan 453002, China
| | - Chang-Hong Wang
- The Second Affiliated Hospital of Xinxiang Medical University and Henan Province Mental Hospital, Xinxiang, Henan 453002, China
| |
Collapse
|
28
|
Caldwell HK, Aulino EA, Rodriguez KM, Witchey SK, Yaw AM. Social Context, Stress, Neuropsychiatric Disorders, and the Vasopressin 1b Receptor. Front Neurosci 2017; 11:567. [PMID: 29085277 PMCID: PMC5650633 DOI: 10.3389/fnins.2017.00567] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/27/2017] [Indexed: 01/28/2023] Open
Abstract
The arginine vasopressin 1b receptor (Avpr1b) is involved in the modulation of a variety of behaviors and is an important part of the mammalian hormonal stress axis. The Avpr1b is prominent in hippocampal CA2 pyramidal cells and in the anterior pituitary corticotrophs. Decades of research on this receptor has demonstrated its importance to the modulation of social recognition memory, social forms of aggression, and modulation of the hypothalamic-pituitary-adrenal axis, particularly under conditions of acute stress. Further, work in humans suggests that the Avpr1b may play a role in human neuropsychiatric disorders and its modulation may have therapeutic potential. This paper reviews what is known about the role of the Avpr1b in the context of social behaviors, the stress axis, and human neuropsychiatric disorders. Further, possible mechanisms for how Avpr1b activation within the hippocampus vs. Avpr1b activation within anterior pituitary may interact with one another to affect behavioral output are proposed.
Collapse
Affiliation(s)
- Heather K Caldwell
- Laboratory of Neuroendocrinology and Behavior, Department of Biological Sciences Kent State University, Kent, OH, United States.,School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Elizabeth A Aulino
- Laboratory of Neuroendocrinology and Behavior, Department of Biological Sciences Kent State University, Kent, OH, United States
| | - Karla M Rodriguez
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Shannah K Witchey
- Laboratory of Neuroendocrinology and Behavior, Department of Biological Sciences Kent State University, Kent, OH, United States
| | - Alexandra M Yaw
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| |
Collapse
|
29
|
Li J, Tang L, Wang Y, Li F, Bao M, Xiang J, Lei D, Tang B. Genetic Associations and Interactions Between the NR3C1 (GR) and NR3C2 (MR) Genes and Aggressive Behavior in a Central South Chinese Han Population. Genet Test Mol Biomarkers 2017; 21:497-505. [PMID: 28686058 DOI: 10.1089/gtmb.2016.0417] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Jianming Li
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Neuroscience Research Center, Changsha Medical University, Changsha, Hunan, China
- Department of Anatomy, Changsha Medical University, Changsha, Hunan, China
| | - Liang Tang
- Neuroscience Research Center, Changsha Medical University, Changsha, Hunan, China
- Department of Anatomy, Changsha Medical University, Changsha, Hunan, China
| | - Yan Wang
- Neuroscience Research Center, Changsha Medical University, Changsha, Hunan, China
- School of Basic Medical Science, Changsha Medical University, Changsha, Hunan, China
| | - Fang Li
- Neuroscience Research Center, Changsha Medical University, Changsha, Hunan, China
- Department of Anatomy, Changsha Medical University, Changsha, Hunan, China
| | - Meihua Bao
- Neuroscience Research Center, Changsha Medical University, Changsha, Hunan, China
- Department of Anatomy, Changsha Medical University, Changsha, Hunan, China
| | - Ju Xiang
- Neuroscience Research Center, Changsha Medical University, Changsha, Hunan, China
- Department of Anatomy, Changsha Medical University, Changsha, Hunan, China
| | - Deliang Lei
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Anatomy and Neurobiology, Central South University Xiangya Medical School, Changsha, Hunan, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
30
|
Taylor JH, Intorre AA, French JA. Vasopressin and Oxytocin Reduce Food Sharing Behavior in Male, but Not Female Marmosets in Family Groups. Front Endocrinol (Lausanne) 2017; 8:181. [PMID: 28798724 PMCID: PMC5529352 DOI: 10.3389/fendo.2017.00181] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 07/10/2017] [Indexed: 12/02/2022] Open
Abstract
Oxytocin (OT) is critical for lactation and maternal care, but OT and the related nonapeptide vasopressin are important for caregiving behaviors in fathers and alloparents as well. This experiment tested the effects of vasopressin and OT on food sharing in marmoset families. We treated caregivers (parents, siblings) with intranasal vasopressin, OT, or saline, and then paired them with the youngest marmoset in the family. Caregivers were given preferred food, and then observed for food sharing and aggressive behavior with young marmosets. OT reduced food sharing from male alloparents to youngest siblings, and fathers that received vasopressin refused to share food with their youngest offspring more often than when treated with OT. Vasopressin increased aggressive vocalizations directed toward potential food recipients in all classes of caregivers. These results indicate that vasopressin and OT do not always enhance prosocial behavior: modulation of food sharing depends on both sex and parental status.
Collapse
Affiliation(s)
- Jack H. Taylor
- Department of Psychology, University of Nebraska at Omaha, Omaha, NE, United States
- Callitrichid Research Center, Omaha, NE, United States
| | - Allison A. Intorre
- Department of Psychology, University of Nebraska at Omaha, Omaha, NE, United States
- Callitrichid Research Center, Omaha, NE, United States
| | - Jeffrey A. French
- Department of Psychology, University of Nebraska at Omaha, Omaha, NE, United States
- Callitrichid Research Center, Omaha, NE, United States
- Department of Biology, University of Nebraska at Omaha, Omaha, NE, United States
| |
Collapse
|
31
|
The Role of the Oxytocin/Arginine Vasopressin System in Animal Models of Autism Spectrum Disorder. ADVANCES IN ANATOMY EMBRYOLOGY AND CELL BIOLOGY 2017; 224:135-158. [DOI: 10.1007/978-3-319-52498-6_8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
32
|
Abstract
For many, the terms oxytocin and vasopressin immediately evoke images of animals interacting with one another, as both of these neuropeptides have been implicated as being part of the neurochemical "glue" that socially binds animals. However, social environments and social interactions are complex and include behaviors that bring animals together as well as behaviors that keep animals apart. It is at the intersection of social context, social experience, and an individual's sex that oxytocin and vasopressin act to modulate social behavior and social cognition. In this review, this complexity will be explored across mammalian species, with a focus on social memory, cooperative behaviors, and competitive behaviors. Implications for humans as well as future directions will also be considered.
Collapse
Affiliation(s)
- Heather K Caldwell
- 1 Laboratory of Neuroendocrinology and Behavior, Department of Biological Sciences and School of Biomedical Sciences, Kent State University, Kent, OH, USA
| |
Collapse
|
33
|
Terranova JI, Ferris CF, Albers HE. Sex Differences in the Regulation of Offensive Aggression and Dominance by Arginine-Vasopressin. Front Endocrinol (Lausanne) 2017; 8:308. [PMID: 29184535 PMCID: PMC5694440 DOI: 10.3389/fendo.2017.00308] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 10/23/2017] [Indexed: 02/01/2023] Open
Abstract
Arginine-vasopressin (AVP) plays a critical role in the regulation of offensive aggression and social status in mammals. AVP is found in an extensive neural network in the brain. Here, we discuss the role of AVP in the regulation of aggression in the limbic system with an emphasis on the critical role of hypothalamic AVP in the control of aggression. In males, activation of AVP V1a receptors (V1aRs) in the hypothalamus stimulates offensive aggression, while in females activation of V1aRs inhibits aggression. Serotonin (5-HT) also acts within the hypothalamus to modulate the effects of AVP on aggression in a sex-dependent manner. Activation of 5-HT1a receptors (5-HT1aRs) inhibits aggression in males and stimulates aggression in females. There are also striking sex differences in the mechanisms underlying the acquisition of dominance. In males, the acquisition of dominance is associated with the activation of AVP-containing neurons in the hypothalamus. By contrast, in females, the acquisition of dominance is associated with the activation of 5-HT-containing neurons in the dorsal raphe. AVP and 5-HT also play critical roles in the regulation of a form of social communication that is important for the maintenance of dominance relationships. In both male and female hamsters, AVP acts via V1aRs in the hypothalamus, as well as in other limbic structures, to communicate social status through the stimulation of a form of scent marking called flank marking. 5-HT acts on 5-HT1aRs as well as other 5-HT receptors within the hypothalamus to inhibit flank marking induced by AVP in both males and females. Interestingly, while AVP and 5-HT influence the expression of aggression in opposite ways in males and females, there are no sex differences in the effects of AVP and 5-HT on the expression of social communication. Given the profound sex differences in the incidence of many psychiatric disorders and the increasing evidence for a relationship between aggressiveness/dominance and the susceptibility to these disorders, understanding the neural regulation of aggression and social status will have significant import for translational studies.
Collapse
Affiliation(s)
- Joseph I. Terranova
- Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Craig F. Ferris
- Department of Psychology, Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States
| | - H. Elliott Albers
- Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, GA, United States
- *Correspondence: H. Elliott Albers,
| |
Collapse
|
34
|
Witchey SK, Stevenson EL, Caldwell HK. Genotypic differences in intruder-evoked immediate early gene activation in male, but not female, vasopressin 1b receptor knockout mice. BMC Neurosci 2016; 17:75. [PMID: 27881080 PMCID: PMC5122005 DOI: 10.1186/s12868-016-0310-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 11/16/2016] [Indexed: 12/27/2022] Open
Abstract
Background The neuropeptide arginine vasopressin (Avp) modulates social behaviors via its two centrally expressed receptors, the Avp 1a receptor and the Avp 1b receptor (Avpr1b). Recent work suggests that, at least in mice, Avp signaling through Avpr1b within the CA2 region of the hippocampus is critical for normal aggressive behaviors and social recognition memory. However, this brain area is just one part of a larger neural circuit that is likely to be impacted in Avpr1b knockout (−/−) mice. To identify other brain areas that are affected by altered Avpr1b signaling, genotypic differences in immediate early gene activation, i.e. c-FOS and early growth response factor 1 (EGR-1), were quantified using immunocytochemistry following a single exposure to an intruder. Results In females, no genotypic differences in intruder-evoked c-FOS or EGR-1 immunoreactivity were observed in any of the brain areas measured. In males, while there were no intruder-evoked genotypic differences in c-FOS immunoreactivity, genotypic differences were observed in EGR-1 immunoreactivity within the ventral bed nucleus of the stria terminalis and the anterior hypothalamus; with Avpr1b −/− males having less EGR-1 immunoreactivity in these regions than controls. Conclusions These data are the first to identify specific brain areas that may be a part of a neural circuit that includes Avpr1b-expressing cells in the CA2 region of the hippocampus. It is thought that this circuit, when working properly, plays a role in how an animal evaluates its social context. Electronic supplementary material The online version of this article (doi:10.1186/s12868-016-0310-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shannah K Witchey
- Laboratory of Neuroendocrinology and Behavior, Department of Biological Sciences, Kent State University, 256 Cunningham Hall, Kent, OH, 44242, USA
| | - Erica L Stevenson
- Laboratory of Neuroendocrinology and Behavior, Department of Biological Sciences, Kent State University, 256 Cunningham Hall, Kent, OH, 44242, USA.,Laboratory of Neuroendocrinology and Behavior, School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | - Heather K Caldwell
- Laboratory of Neuroendocrinology and Behavior, Department of Biological Sciences, Kent State University, 256 Cunningham Hall, Kent, OH, 44242, USA. .,Laboratory of Neuroendocrinology and Behavior, School of Biomedical Sciences, Kent State University, Kent, OH, USA.
| |
Collapse
|
35
|
Freudenberg F, Carreño Gutierrez H, Post AM, Reif A, Norton WHJ. Aggression in non-human vertebrates: Genetic mechanisms and molecular pathways. Am J Med Genet B Neuropsychiatr Genet 2016; 171:603-40. [PMID: 26284957 DOI: 10.1002/ajmg.b.32358] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/28/2015] [Indexed: 11/07/2022]
Abstract
Aggression is an adaptive behavioral trait that is important for the establishment of social hierarchies and competition for mating partners, food, and territories. While a certain level of aggression can be beneficial for the survival of an individual or species, abnormal aggression levels can be detrimental. Abnormal aggression is commonly found in human patients with psychiatric disorders. The predisposition to aggression is influenced by a combination of environmental and genetic factors and a large number of genes have been associated with aggression in both human and animal studies. In this review, we compare and contrast aggression studies in zebrafish and mouse. We present gene ontology and pathway analyses of genes linked to aggression and discuss the molecular pathways that underpin agonistic behavior in these species. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Florian Freudenberg
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Frankfurt am Main, Germany
| | | | - Antonia M Post
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Frankfurt am Main, Germany
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital of Frankfurt, Frankfurt am Main, Germany
| | - William H J Norton
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester, UK
| |
Collapse
|
36
|
Bayerl DS, Kaczmarek V, Jurek B, van den Burg EH, Neumann ID, Gaßner BM, Klampfl SM, Bosch OJ. Antagonism of V1b receptors promotes maternal motivation to retrieve pups in the MPOA and impairs pup-directed behavior during maternal defense in the mpBNST of lactating rats. Horm Behav 2016; 79:18-27. [PMID: 26747375 DOI: 10.1016/j.yhbeh.2015.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 12/26/2015] [Accepted: 12/29/2015] [Indexed: 01/30/2023]
Abstract
Recent studies using V1b receptor (V1bR) knockout mice or central pharmacological manipulations in lactating rats highlighted the influence of this receptor for maternal behavior. However, its role in specific brain sites known to be important for maternal behavior has not been investigated to date. In the present study, we reveal that V1bR mRNA (qPCR) and protein levels (Western blot) within either the medial preoptic area (MPOA) or the medial-posterior part of the bed nucleus of the stria terminalis (mpBNST) did not differ between virgin and lactating rats. Furthermore, we characterized the effects of V1bR blockade via bilateral injections of the receptor subtype-specific antagonist SSR149415 within the MPOA or the mpBNST on maternal behavior (maternal care under non-stress and stress conditions, maternal motivation to retrieve pups in a novel environment, maternal aggression) and anxiety-related behavior in lactating rats. Blocking V1bR within the MPOA increased pup retrieval, whereas within the mpBNST it decreased pup-directed behavior, specifically licking/grooming the pups, during the maternal defense test. In addition, immediately after termination of the maternal defense test, V1bR antagonism in both brain regions reduced nursing, particularly arched back nursing. Anxiety-related behavior was not affected by V1bR antagonism in either brain region. In conclusion our data indicate that V1bR antagonism significantly modulates different aspects of maternal behavior in a brain region-dependent manner.
Collapse
Affiliation(s)
- Doris S Bayerl
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, 93053 Regensburg, Germany.
| | - Veronika Kaczmarek
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, 93053 Regensburg, Germany.
| | - Benjamin Jurek
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, 93053 Regensburg, Germany.
| | - Erwin H van den Burg
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, 93053 Regensburg, Germany.
| | - Inga D Neumann
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, 93053 Regensburg, Germany.
| | - Barbara M Gaßner
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, 93053 Regensburg, Germany.
| | - Stefanie M Klampfl
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, 93053 Regensburg, Germany.
| | - Oliver J Bosch
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, 93053 Regensburg, Germany.
| |
Collapse
|
37
|
Caldwell HK, Albers HE. Oxytocin, Vasopressin, and the Motivational Forces that Drive Social Behaviors. Curr Top Behav Neurosci 2016; 27:51-103. [PMID: 26472550 DOI: 10.1007/7854_2015_390] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The motivation to engage in social behaviors is influenced by past experience and internal state, but also depends on the behavior of other animals. Across species, the oxytocin (Oxt) and vasopressin (Avp) systems have consistently been linked to the modulation of motivated social behaviors. However, how they interact with other systems, such as the mesolimbic dopamine system, remains understudied. Further, while the neurobiological mechanisms that regulate prosocial/cooperative behaviors have been extensively examined, far less is understood about competitive behaviors, particularly in females. In this chapter, we highlight the specific contributions of Oxt and Avp to several cooperative and competitive behaviors and discuss their relevance to the concept of social motivation across species, including humans. Further, we discuss the implications for neuropsychiatric diseases and suggest future areas of investigation.
Collapse
|
38
|
The neurobiology of offensive aggression: Revealing a modular view. Physiol Behav 2015; 146:111-27. [DOI: 10.1016/j.physbeh.2015.04.040] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 04/21/2015] [Accepted: 04/22/2015] [Indexed: 02/03/2023]
|
39
|
Abstract
Aggression and violence represent a significant public health concern and a clinical challenge for the mental healthcare provider. A great deal has been revealed regarding the neurobiology of violence and aggression, and an integration of this body of knowledge will ultimately serve to advance clinical diagnostics and therapeutic interventions. We will review here the latest findings regarding the neurobiology of aggression and violence. First, we will introduce the construct of aggression, with a focus on issues related to its heterogeneity, as well as the importance of refining the aggression phenotype in order to reduce pathophysiologic variability. Next we will examine the neuroanatomy of aggression and violence, focusing on regional volumes, functional studies, and interregional connectivity. Significant emphasis will be on the amygdala, as well as amygdala-frontal circuitry. Then we will turn our attention to the neurochemistry and molecular genetics of aggression and violence, examining the extensive findings on the serotonergic system, as well as the growing literature on the dopaminergic and vasopressinergic systems. We will also address the contribution of steroid hormones, namely, cortisol and testosterone. Finally, we will summarize these findings with a focus on reconciling inconsistencies and potential clinical implications; and, then we will suggest areas of focus for future directions in the field.
Collapse
|
40
|
Role of the vasopressin 1b receptor in rodent aggressive behavior and synaptic plasticity in hippocampal area CA2. Mol Psychiatry 2015; 20:490-9. [PMID: 24863146 PMCID: PMC4562468 DOI: 10.1038/mp.2014.47] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 03/07/2014] [Accepted: 04/16/2014] [Indexed: 01/31/2023]
Abstract
The vasopressin 1b receptor (Avpr1b) is critical for social memory and social aggression in rodents, yet little is known about its specific roles in these behaviors. Some clues to Avpr1b function can be gained from its profile of expression in the brain, which is largely limited to the pyramidal neurons of the CA2 region of the hippocampus, and from experiments showing that inactivation of the gene or antagonism of the receptor leads to a reduction in social aggression. Here we show that partial replacement of the Avpr1b through lentiviral delivery into the dorsal CA2 region restored the probability of socially motivated attack behavior in total Avpr1b knockout mice, without altering anxiety-like behaviors. To further explore the role of the Avpr1b in this hippocampal region, we examined the effects of Avpr1b agonists on pyramidal neurons in mouse and rat hippocampal slices. We found that selective Avpr1b agonists induced significant potentiation of excitatory synaptic responses in CA2, but not in CA1 or in slices from Avpr1b knockout mice. In a way that is mechanistically very similar to synaptic potentiation induced by oxytocin, Avpr1b agonist-induced potentiation of CA2 synapses relies on NMDA (N-methyl-D-aspartic acid) receptor activation, calcium and calcium/calmodulin-dependent protein kinase II activity, but not on cAMP-dependent protein kinase activity or presynaptic mechanisms. Our data indicate that the hippocampal CA2 is important for attacking in response to a male intruder and that the Avpr1b, likely through its role in regulating CA2 synaptic plasticity, is a necessary mediator.
Collapse
|
41
|
Jones LJ, Norton WH. Using zebrafish to uncover the genetic and neural basis of aggression, a frequent comorbid symptom of psychiatric disorders. Behav Brain Res 2015; 276:171-80. [DOI: 10.1016/j.bbr.2014.05.055] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 05/23/2014] [Accepted: 05/26/2014] [Indexed: 12/31/2022]
|
42
|
Zimmermann-Peruzatto JM, Lazzari VM, de Moura AC, Almeida S, Giovenardi M. Examining the Role of Vasopressin in the Modulation of Parental and Sexual Behaviors. Front Psychiatry 2015; 6:130. [PMID: 26441691 PMCID: PMC4585274 DOI: 10.3389/fpsyt.2015.00130] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Accepted: 09/04/2015] [Indexed: 11/13/2022] Open
Abstract
Vasopressin (VP) and VP-like neuropeptides are evolutionarily stable peptides found in all vertebrate species. In non-mammalian vertebrates, vasotocin (VT) plays a role similar to mammalian VP, whereas mesotocin and isotocin are functionally similar to mammalian oxytocin (OT). Here, we review the involvement of VP in brain circuits, synaptic plasticity, evolution, and function, highlighting the role of VP in social behavior. In all studied species, VP is encoded on chromosome 20p13, and in mammals, VP is produced in specific hypothalamic nuclei and released by the posterior pituitary. The role of VP is mediated by the stimulation of the V1a, V1b, and V2 receptors as well as the oxytocinergic and purinergic receptors. VT and VP functions are usually related to osmotic and cardiovascular homeostasis when acting peripherally. However, these neuropeptides are also critically involved in the central modulation of social behavior displays, such as pairing recognition, pair-bonding, social memory, sexual behavior, parental care, and maternal and aggressive behavior. Evidence suggests that these effects are primarily mediated by V1a receptor in specific brain circuits that provide important information for the onset and control of social behaviors in normal and pathological conditions.
Collapse
Affiliation(s)
- Josi Maria Zimmermann-Peruzatto
- Programa de Pós-Graduação em Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS) , Porto Alegre , Brazil
| | - Virgínia Meneghini Lazzari
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA) , Porto Alegre , Brazil
| | - Ana Carolina de Moura
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA) , Porto Alegre , Brazil
| | - Silvana Almeida
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA) , Porto Alegre , Brazil
| | - Márcia Giovenardi
- Programa de Pós-Graduação em Fisiologia, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS) , Porto Alegre , Brazil ; Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA) , Porto Alegre , Brazil
| |
Collapse
|
43
|
Bayerl DS, Klampfl SM, Bosch OJ. Central V1b receptor antagonism in lactating rats: impairment of maternal care but not of maternal aggression. J Neuroendocrinol 2014; 26:918-26. [PMID: 25283607 DOI: 10.1111/jne.12226] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 09/30/2014] [Accepted: 09/30/2014] [Indexed: 01/22/2023]
Abstract
Maternal behaviour in rodents is mediated by the central oxytocin and vasopressin systems, amongst others. The role of vasopressin, acting via the V1a receptor (V1aR), on maternal care and maternal aggression has recently been described. However, a potential involvement of the V1b receptor (V1bR) in maternal behaviour has only been demonstrated in knockout mice. The present study aimed to examine the effects of central pharmacological manipulation of the V1bR on maternal behaviour in lactating Wistar rats. On pregnancy day 18, female rats were implanted with a guide cannula targeting the lateral ventricle. After parturition, dams received an acute central infusion of a specific V1bR agonist (d[Leu4,Lys8]VP) or V1bR antagonist (SSR149415) once daily, followed by observations of maternal care [lactation day (LD) 1], maternal motivation in the pup retrieval test (LD 2), anxiety-related behaviour on the elevated plus-maze (LD 3) and maternal aggression in the maternal defence test followed by maternal care monitoring (LD 4). Our data demonstrate that, under nonstress conditions, the V1bR antagonist decreased the occurrence of both nursing and mother-pup interaction, whereas the V1bR agonist did not affect either parameter. Under stress conditions (i.e. after the maternal defence test), mother-pup interaction was decreased by infusion of the V1bR antagonist. During the maternal defence test, neither treatment affected aggressive or non-aggressive behaviour. Finally, neither treatment altered maternal motivation or anxiety. In conclusion, central V1bR antagonism modulates aspects of maternal care but not of maternal aggression or maternal motivation in lactating rats. These findings further extend our knowledge on the vasopressin system as a vital mediator of maternal behaviour.
Collapse
Affiliation(s)
- D S Bayerl
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, Regensburg, Germany
| | | | | |
Collapse
|
44
|
Abstract
Suicide ranks among the leading causes of death around the world and takes a heavy emotional and public health toll on most societies. Both distal and proximal factors contribute to suicidal behaviour. Distal factors - such as familial and genetic predisposition, as well as early-life adversity - increase the lifetime risk of suicide. They alter responses to stress and other processes through epigenetic modification of genes and associated changes in gene expression, and through the regulation of emotional and behavioural traits. Proximal factors are associated with the precipitation of a suicidal event and include alterations in key neurotransmitter systems, inflammatory changes and glial dysfunction in the brain. This Review explores the key molecular changes that are associated with suicidality and discusses some promising avenues for future research.
Collapse
|
45
|
Stevenson EL, Caldwell HK. Lesions to the CA2 region of the hippocampus impair social memory in mice. Eur J Neurosci 2014; 40:3294-301. [PMID: 25131412 DOI: 10.1111/ejn.12689] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/29/2014] [Accepted: 07/11/2014] [Indexed: 12/26/2022]
Abstract
The function of the CA2 region of the hippocampus is poorly understood. Although the CA1 and CA3 regions have been extensively studied, for years the CA2 region has primarily been viewed as a linking area between the two. However, the CA2 region is known to have distinct neurochemical and structural features that are different from the other parts of the hippocampus and in recent years it has been suggested that the CA2 region may play a role in the formation and/or recall of olfactory-based memories needed for normal social behavior. Although this hypothesis has been supported by hippocampal lesion studies that have included the CA2 region, no studies have attempted to specifically lesion the CA2 region of the hippocampus in mice to determine the effects on social recognition memory and olfaction. To fill this knowledge gap, we sought to perform excitotoxic N-methyl-D-aspartate lesions of the CA2 region and determine the effects on social recognition memory. We predicted that lesions of the CA2 region would impair social recognition memory. We then went on to test olfaction in CA2-lesioned mice, as social memory requires a functional olfactory system. Consistent with our prediction, we found that CA2-lesioned animals had impaired social recognition. These findings are significant because they confirmed that the CA2 region of the hippocampus is a part of the neural circuitry that regulates social recognition memory, which may have implications for our understanding of the neural regulation of social behavior across species.
Collapse
Affiliation(s)
- Erica L Stevenson
- Laboratory of Neuroendocrinology and Behavior, Department of Biological Sciences and the School of Biomedical Sciences, Kent State University, 121 Cunningham Hall, Kent, OH, 44242, USA
| | | |
Collapse
|
46
|
Bowen MT, Dass SAH, Booth J, Suraev A, Vyas A, McGregor IS. Active coping toward predatory stress is associated with lower corticosterone and progesterone plasma levels and decreased methylation in the medial amygdala vasopressin system. Horm Behav 2014; 66:561-6. [PMID: 25127982 DOI: 10.1016/j.yhbeh.2014.08.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 08/04/2014] [Accepted: 08/06/2014] [Indexed: 01/30/2023]
Abstract
An active coping style displayed under stress - which involves proactive investigatory responses toward environmental threats - has been associated with reduced vulnerability to psychiatric illness. However, the neurobiological determinants of coping styles are not well understood. When rats are exposed to a naturalistic stressor (cat fur) in a group, some individuals in the group show robust active investigation of the stimulus while others show a passive response involving retreat, immobility and close aggregation with conspecifics. Here we explored endocrine and epigenetic correlates of these contrasting coping styles. Male Wistar rats (n=48) were exposed to cat fur in groups of 4 and the passive and active responders were identified and assessed for endocrine and epigenetic differences. Three days after the final cat fur exposure, active responders had substantially lower plasma levels of corticosterone and progesterone than passive responders. Plasma and testicular testosterone levels did not differ between active and passive responders. Active responders had markedly less methylation of the AVP CGCG promoter region located at base 4970 in the posterodorsal region of the medial amygdala but did not differ in the methylation status of the CCGG sequence located at base 2243. This is in agreement with prior research suggesting that AVP and progesterone act in opposition within the medial amygdala to modulate stress-related behaviors. The present study reports striking endocrine and epigenetic differences between active and passive responders, providing insight into potential systems involved in the manifestation of differing coping styles.
Collapse
Affiliation(s)
- Michael T Bowen
- School of Psychology, University of Sydney, Sydney, NSW, Australia
| | - Shantala A Hari Dass
- School of Biological Sciences, Nanyang Technological University, Singapore, Republic of Singapore
| | - Jessica Booth
- School of Psychology, University of Sydney, Sydney, NSW, Australia
| | - Anastasia Suraev
- School of Psychology, University of Sydney, Sydney, NSW, Australia
| | - Ajai Vyas
- School of Biological Sciences, Nanyang Technological University, Singapore, Republic of Singapore
| | - Iain S McGregor
- School of Psychology, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
47
|
Morrison TR, Melloni RH. The role of serotonin, vasopressin, and serotonin/vasopressin interactions in aggressive behavior. Curr Top Behav Neurosci 2014; 17:189-228. [PMID: 24496652 DOI: 10.1007/7854_2014_283] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Aggression control has been investigated across species and is centrally mediated within various brain regions by several neural systems that interact at different levels. The debate over the degree to which any one system or region affects aggressive responding, or any behavior for that matter, in some senses is arbitrary considering the plastic and adaptive properties of the central nervous system. Nevertheless, from the reductionist point of view, the compartmentalization of evolutionarily maladaptive behaviors to specific regions and systems of the brain is necessary for the advancement of clinical treatments (e.g., pharmaceutical) and novel therapeutic methods (e.g., deep brain stimulation). The general purpose of this chapter is to examine the confluence of two such systems, and how their functional interaction affects aggressive behavior. Specifically, the influence of the serotonin (5HT) and arginine vasopressin (AVP) neural systems on the control of aggressive behavior will be examined individually and together to provide a context by which the understanding of aggression modulation can be expanded from seemingly parallel neuromodulatory mechanisms, to a single and highly interactive system of aggression control.
Collapse
Affiliation(s)
- Thomas R Morrison
- Program in Behavioral Neuroscience, Department of Psychology, Northeastern University, 125 Nightingale Hall, 360 Huntington Ave, Boston, MA, 02155, USA,
| | | |
Collapse
|
48
|
Trojan Genes or Transparent Genomes? Sexual Selection and Potential Impacts of Genetically Modified Animals in Natural Ecosystems. Evol Biol 2013. [DOI: 10.1007/s11692-013-9268-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
49
|
Bosch OJ. Maternal aggression in rodents: brain oxytocin and vasopressin mediate pup defence. Philos Trans R Soc Lond B Biol Sci 2013; 368:20130085. [PMID: 24167315 DOI: 10.1098/rstb.2013.0085] [Citation(s) in RCA: 128] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The most significant social behaviour of the lactating mother is maternal behaviour, which comprises maternal care and maternal aggression (MA). The latter is a protective behaviour of the mother serving to defend the offspring against a potentially dangerous intruder. The extent to which the mother shows aggressive behaviour depends on extrinsic and intrinsic factors, as we have learned from studies in laboratory rodents. Among the extrinsic factors are the pups' presence and age, as well as the intruders' sex and age. With respect to intrinsic factors, the mothers' innate anxiety and the prosocial brain neuropeptides oxytocin (OXT) and arginine vasopressin (AVP) play important roles. While OXT is well known as a maternal neuropeptide, AVP has only recently been described in this context. The increased activities of these neuropeptides in lactation are the result of remarkable brain adaptations peripartum and are a prerequisite for the mother to become maternal. Consequently, OXT and AVP are significantly involved in mediating the fine-tuned regulation of MA depending on the brain regions. Importantly, both neuropeptides are also modulators of anxiety, which determines the extent of MA. This review provides a detailed overview of the role of OXT and AVP in MA and the link to anxiety.
Collapse
Affiliation(s)
- Oliver J Bosch
- Department of Behavioural and Molecular Neurobiology, University of Regensburg, , Regensburg 93040, Germany
| |
Collapse
|
50
|
Cui Z, Gerfen CR, Young WS. Hypothalamic and other connections with dorsal CA2 area of the mouse hippocampus. J Comp Neurol 2013; 521:1844-66. [PMID: 23172108 DOI: 10.1002/cne.23263] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 06/22/2012] [Accepted: 11/06/2012] [Indexed: 11/08/2022]
Abstract
The CA2 area is an important, although relatively unexplored, component of the hippocampus. We used various tracers to provide a comprehensive analysis of CA2 connections in C57BL/6J mice. Using various adeno-associated viruses that express fluorescent proteins, we found a vasopressinergic projection from the paraventricular nuclei of the hypothalamus (PVN) to the CA2 as well as a projection from pyramidal neurons of the CA2 to the supramammillary nuclei. These projections were confirmed by retrograde tracing. As expected, we observed CA2 afferent projections from neurons in ipsilateral entorhinal cortical layer II as well as from bilateral dorsal CA2 and CA3 using retrograde tracers. Additionally, we saw CA2 neuronal input from bilateral medial septal nuclei, vertical and horizontal limbs of the nucleus of diagonal band of Broca, supramammillary nuclei (SUM), and median raphe nucleus. Dorsal CA2 injections of adeno-associated virus expressing green fluorescent protein revealed axonal projections primarily to dorsal CA1, CA2, and CA3 bilaterally. No projection was detected to the entorhinal cortex from the dorsal CA2. These results are consistent with recent observations that the dorsal CA2 forms disynaptic connections with the entorhinal cortex to influence dynamic memory processing. Mouse dorsal CA2 neurons send bilateral projections to the medial and lateral septal nuclei, vertical and horizontal limbs of the diagonal band of Broca, and SUM. Novel connections from the PVN and to the SUM suggest important regulatory roles for CA2 in mediating social and emotional input for memory processing.
Collapse
Affiliation(s)
- Zhenzhong Cui
- Section on Neural Gene Expression, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland 20892, USA
| | | | | |
Collapse
|