1
|
Makkar J, Flores J, Matich M, Duong TT, Thompson SM, Du Y, Busch I, Phan QM, Wang Q, Delevich K, Broughton-Neiswanger L, Driskell IM, Driskell RR. Deep Hair Phenomics: Implications in Endocrinology, Development, and Aging. J Invest Dermatol 2025; 145:800-811.e8. [PMID: 39236901 PMCID: PMC11873809 DOI: 10.1016/j.jid.2024.08.014] [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: 04/02/2024] [Revised: 08/08/2024] [Accepted: 08/11/2024] [Indexed: 09/07/2024]
Abstract
Hair quality is an important indicator of health in humans and other animals. Current approaches to assess hair quality are generally nonquantitative or are low throughput owing to technical limitations of splitting hairs. We developed a deep learning-based computer vision approach for the high-throughput quantification of individual hair fibers at a high resolution. Our innovative computer vision tool can distinguish and extract overlapping fibers for quantification of multivariate features, including length, width, and color, to generate single-hair phenomes of diverse conditions across the lifespan of mice. Using our tool, we explored the effects of hormone signaling, genetic modifications, and aging on hair follicle output. Our analyses revealed hair phenotypes resultant of endocrinological, developmental, and aging-related alterations in the fur coats of mice. These results demonstrate the efficacy of our deep hair phenomics tool for characterizing factors that modulate the hair follicle and developing, to our knowledge, previously unreported diagnostic methods for detecting disease through the hair fiber. Finally, we have generated a searchable, interactive web tool for the exploration of our hair fiber data at skinregeneration.org.
Collapse
Affiliation(s)
- Jasson Makkar
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| | - Jorge Flores
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| | - Mason Matich
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| | - Tommy T Duong
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| | - Sean M Thompson
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| | - Yiqing Du
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| | - Isabelle Busch
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| | - Quan M Phan
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| | - Qing Wang
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Kristen Delevich
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA; Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Liam Broughton-Neiswanger
- Washington Animal Disease Diagnostic Laboratory, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Iwona M Driskell
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| | - Ryan R Driskell
- School of Molecular Biosciences, Washington State University, Pullman, Washington, USA; Center for Reproductive Biology, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA.
| |
Collapse
|
2
|
Chaturvedi SM, Sarafinovska S, Selmanovic D, McCullough KB, Swift RG, Maloney SE, Dougherty JD. Chromosomal and gonadal sex have differing effects on social motivation in mice. Biol Sex Differ 2025; 16:13. [PMID: 39966983 PMCID: PMC11837725 DOI: 10.1186/s13293-025-00690-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/25/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Sex differences in brain development are thought to lead to sex variation in social behavior. Sex differences are fundamentally driven by both gonadal hormones and sex chromosomes, yet little is known about the independent effects of each on social behavior. Further, mouse models of the genetic liability for the neurodevelopmental disorder MYT1L Syndrome have shown sex-specific deficits in social motivation. In this study, we aimed to determine if gonadal hormones or sex chromosomes primarily mediate the sex differences seen in mouse social behavior, both at baseline and in the context of Myt1l haploinsufficiency. METHODS Four-core genotypes (FCG) mice, which uncouple gonadal and chromosomal sex, were crossed with MYT1L heterozygous mice to create eight different groups with unique combinations of sex factors and MYT1L genotype. A total of 131 mice from all eight groups were assayed for activity and social behavior via the open field and social operant paradigms. Measures of social seeking and orienting were analyzed for main effects of chromosome, gonads, and their interactions with Myt1l mutation. RESULTS The FCGxMYT1L cross revealed independent effects of both gonadal and chromosomal sex on activity and social behavior. Specifically, the presence of ovarian hormones led to greater overall activity, social seeking, and social orienting regardless of MYT1L genotype. In contrast, sex chromosomes affected social behavior mainly in the MYT1L heterozygous group, with XX MYT1L mutant mice demonstrating elevated levels of social orienting and seeking compared to XY MYT1L mutant mice. CONCLUSIONS Gonadal and chromosomal sex have independent mechanisms of driving greater social motivation in females. Additionally, genes on the sex chromosomes may interact with neurodevelopmental risk genes to influence sex variation in atypical social behavior.
Collapse
Affiliation(s)
- Sneha M Chaturvedi
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Simona Sarafinovska
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Din Selmanovic
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Katherine B McCullough
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Raylynn G Swift
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Susan E Maloney
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, 63130, USA
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA.
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110, USA.
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, 63130, USA.
| |
Collapse
|
3
|
Shivakumar AB, Mehak SF, Gupta A, Gangadharan G. Medial septal cholinergic neurotransmission is essential for social memory in mice. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111207. [PMID: 39615870 DOI: 10.1016/j.pnpbp.2024.111207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/09/2024] [Accepted: 11/24/2024] [Indexed: 01/29/2025]
Abstract
Social memory, a fundamental component of social behavior, is essential for the recognition and recall of familiar and novel animals/humans which is disrupted in several neuropsychiatric disorders. Although hippocampal circuitry is crucial for social memory, the role of extra-hippocampal regions in this behavior remains elusive. Here, we identified the physiological link between medial septal dependent cholinergic theta oscillations in the hippocampus and social memory behavior. We found that selective ablation of cholinergic neurons in the medial septum impaired social memory in mice, while their sociability and social novelty remained intact. Additionally, these mice showed an attenuation of cholinergic theta oscillations (3-7 Hz) in the hippocampal dorsal CA2 (dCA2) region. Furthermore, enhancing dCA2 theta oscillations by elevating cholinergic signaling using acetylcholinesterase inhibitor rescued social memory deficit. Together, these results indicate that 1) medial septal cholinergic neurons are essential for modulating social memory 2) cholinergic hippocampal theta oscillations contribute to social memory processes.
Collapse
Affiliation(s)
- Apoorva Bettagere Shivakumar
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Sonam Fathima Mehak
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Amritanshu Gupta
- Department of Biomedical Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| | - Gireesh Gangadharan
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India.
| |
Collapse
|
4
|
Chaturvedi SM, Sarafinovska S, Selmanovic D, McCullough KB, Swift RG, Maloney SE, Dougherty JD. Chromosomal and gonadal sex have differing effects on social motivation in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620727. [PMID: 39554131 PMCID: PMC11565840 DOI: 10.1101/2024.10.28.620727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Background Sex differences in brain development are thought to lead to sex variation in social behavior. Sex differences are fundamentally driven by both gonadal (i.e., hormonal) and chromosomal sex, yet little is known about the independent effects of each on social behavior. Further, mouse models of the genetic liability for the neurodevelopmental disorder MYT1L Syndrome have shown sex specific deficits in social motivation. In this study, we aimed to determine if hormonal or chromosomal sex primarily mediate the sex differences seen in mouse social behavior, both at baseline and in the context of Myt1l haploinsufficiency. Methods Four-core genotype (FCG) mice, which uncouple gonadal and chromosomal sex, were crossed with MYT1L heterozygous mice to create eight different groups with unique combinations of sex factors and MYT1L genotype. A total of 131 mice from all eight groups were assayed for activity and social behavior via the open field and social operant paradigms. Measures of social seeking and orienting were analyzed for main effects of chromosome, gonads, and their interactions with Myt1l mutation. Results The FCGxMYT1L cross revealed independent effects of both gonadal and chromosomal sex on activity and social behavior. Specifically, the presence of ovaries, and by extension the presence of ovarian hormones, increased overall activity, social seeking, and social orienting regardless of genotype. In contrast, sex chromosomes affected social behavior mainly in the MYT1L heterozygous group, with XX sex karyotype when combined with MYT1L genotype contributing to increased social orienting and seeking. Conclusions Gonadal and chromosomal sex have independent mechanisms of driving increased social motivation in females. Additionally, sex chromosomes may interact with neurodevelopmental mutations to influence sex variation in atypical social behavior.
Collapse
Affiliation(s)
- Sneha M. Chaturvedi
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Simona Sarafinovska
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Din Selmanovic
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Katherine B. McCullough
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Raylynn G. Swift
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Susan E. Maloney
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Joseph D. Dougherty
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO 63130, USA
| |
Collapse
|
5
|
Furukawa M, Izumo N, Aoki R, Nagashima D, Ishibashi Y, Matsuzaki H. Behavioural changes in young ovariectomized mice via GPR30-dependent serotonergic nervous system. Eur J Neurosci 2024; 60:5658-5670. [PMID: 39189108 DOI: 10.1111/ejn.16516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/05/2024] [Accepted: 08/12/2024] [Indexed: 08/28/2024]
Abstract
Fluctuations in estradiol levels at each stage of life in women are considered one of the causes of mental diseases through their effects on the central nervous system. During menopause, a decrease in estradiol levels has been reported to affect the serotonin nervous system and induce depression-like and anxiety symptoms. However, the regulation of brain and behaviour during childhood and adolescence is poorly understood. Moreover, the role of oestrogen receptors α and β in the regulation of the serotonergic nervous system has been reported, but little is known about the involvement of G protein-coupled receptor 30. Therefore, in this study, we used an ovariectomized childhood mouse model to analyse behaviour and investigate the effects on the serotonin nervous system. We showed that ovariectomy surgery at 4 weeks of age, which is the weaning period, induced a decrease in spontaneous locomotor activity during the active period and a preference for novel mice over familiar mice in the three-chamber social test at 10 weeks of age. In addition, the administration of G-1, a protein-coupled receptor 30 agonist, to ovariectomized mice suppressed spontaneous locomotor activity and the preference for novel mice. Furthermore, we demonstrated that childhood ovariectomy induces increased tryptophan hydroxylase gene expression in the raphe nucleus and increased serotonin release in the amygdaloid nucleus, and administration of G-1 ameliorated these effects. Our study suggests that G protein-coupled receptor 30-mediated regulation of serotonin synthesis is involved in changes in activity and social-cognitive behaviour due to decreased estradiol levels during childhood.
Collapse
Affiliation(s)
- Megumi Furukawa
- Department of Functional Brain Activities, United Graduate School of Child Development, Osaka University, Suita, Japan
- Center for Pharmaceutical Education, Yokohama University of Pharmacy, Yokohama, Japan
- Research Center for Child Mental Development, University of Fukui, Eiheiji, Japan
| | - Nobuo Izumo
- Laboratory of Pharmacotherapy, Yokohama University of Pharmacy, Yokohama, Japan
- General Health Medical Research Center, Yokohama University of Pharmacy, Yokohama, Japan
| | - Ryoken Aoki
- Department of Functional Brain Activities, United Graduate School of Child Development, Osaka University, Suita, Japan
- Center for Pharmaceutical Education, Yokohama University of Pharmacy, Yokohama, Japan
- Research Center for Child Mental Development, University of Fukui, Eiheiji, Japan
| | - Daichi Nagashima
- General Health Medical Research Center, Yokohama University of Pharmacy, Yokohama, Japan
- Laboratory of Clinical Pharmaceutics, Yokohama University of Pharmacy, Yokohama, Japan
| | - Yukiko Ishibashi
- Laboratory of Drug Analysis, Yokohama University of Pharmacy, Yokohama, Japan
| | - Hideo Matsuzaki
- Department of Functional Brain Activities, United Graduate School of Child Development, Osaka University, Suita, Japan
- Research Center for Child Mental Development, University of Fukui, Eiheiji, Japan
| |
Collapse
|
6
|
Xu QW, Larosa A, Wong TP. Roles of AMPA receptors in social behaviors. Front Synaptic Neurosci 2024; 16:1405510. [PMID: 39056071 PMCID: PMC11269240 DOI: 10.3389/fnsyn.2024.1405510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
As a crucial player in excitatory synaptic transmission, AMPA receptors (AMPARs) contribute to the formation, regulation, and expression of social behaviors. AMPAR modifications have been associated with naturalistic social behaviors, such as aggression, sociability, and social memory, but are also noted in brain diseases featuring impaired social behavior. Understanding the role of AMPARs in social behaviors is timely to reveal therapeutic targets for treating social impairment in disorders, such as autism spectrum disorder and schizophrenia. In this review, we will discuss the contribution of the molecular composition, function, and plasticity of AMPARs to social behaviors. The impact of targeting AMPARs in treating brain disorders will also be discussed.
Collapse
Affiliation(s)
- Qi Wei Xu
- Douglas Hospital Research Centre, Montreal, QC, Canada
| | - Amanda Larosa
- Douglas Hospital Research Centre, Montreal, QC, Canada
| | - Tak Pan Wong
- Douglas Hospital Research Centre, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
7
|
EFSA Panel on Contaminants in the Food Chain (CONTAM), Schrenk D, Bignami M, Bodin L, Chipman JK, del Mazo J, Grasl‐Kraupp B, Hogstrand C, Hoogenboom L(R, Leblanc J, Nebbia CS, Nielsen E, Ntzani E, Petersen A, Sand S, Schwerdtle T, Wallace H, Benford D, Hart A, Schroeder H, Rose M, Vrijheid M, Kouloura E, Bordajandi LR, Riolo F, Vleminckx C. Update of the scientific opinion on tetrabromobisphenol A (TBBPA) and its derivatives in food. EFSA J 2024; 22:e8859. [PMID: 39010865 PMCID: PMC11247339 DOI: 10.2903/j.efsa.2024.8859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024] Open
Abstract
The European Commission asked EFSA to update its 2011 risk assessment on tetrabromobisphenol A (TBBPA) and five derivatives in food. Neurotoxicity and carcinogenicity were considered as the critical effects of TBBPA in rodent studies. The available evidence indicates that the carcinogenicity of TBBPA occurs via non-genotoxic mechanisms. Taking into account the new data, the CONTAM Panel considered it appropriate to set a tolerable daily intake (TDI). Based on decreased interest in social interaction in male mice, a lowest observed adverse effect level (LOAEL) of 0.2 mg/kg body weight (bw) per day was identified and selected as the reference point for the risk characterisation. Applying the default uncertainty factor of 100 for inter- and intraspecies variability, and a factor of 3 to extrapolate from the LOAEL to NOAEL, a TDI for TBBPA of 0.7 μg/kg bw per day was established. Around 2100 analytical results for TBBPA in food were used to estimate dietary exposure for the European population. The most important contributors to the chronic dietary LB exposure to TBBPA were fish and seafood, meat and meat products and milk and dairy products. The exposure estimates to TBBPA were all below the TDI, including those estimated for breastfed and formula-fed infants. Accounting for the uncertainties affecting the assessment, the CONTAM Panel concluded with 90%-95% certainty that the current dietary exposure to TBBPA does not raise a health concern for any of the population groups considered. There were insufficient data on the toxicity of any of the TBBPA derivatives to derive reference points, or to allow a comparison with TBBPA that would support assignment to an assessment group for the purposes of combined risk assessment.
Collapse
|
8
|
McGovern DJ, Ly A, Ecton KL, Huynh DT, Prévost ED, Gonzalez SC, McNulty CJ, Rau AR, Hentges ST, Daigle TL, Tasic B, Baratta MV, Root DH. Ventral tegmental area glutamate neurons mediate nonassociative consequences of stress. Mol Psychiatry 2024; 29:1671-1682. [PMID: 36437312 PMCID: PMC10375863 DOI: 10.1038/s41380-022-01858-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/29/2022]
Abstract
Exposure to trauma is a risk factor for the development of a number of mood disorders, and may enhance vulnerability to future adverse life events. Recent data demonstrate that ventral tegmental area (VTA) neurons expressing the vesicular glutamate transporter 2 (VGluT2) signal and causally contribute to behaviors that involve aversive or threatening stimuli. However, it is unknown whether VTA VGluT2 neurons regulate transsituational outcomes of stress and whether these neurons are sensitive to stressor controllability. This work adapted an operant mouse paradigm to examine the impact of stressor controllability on VTA VGluT2 neuron function as well as the role of VTA VGluT2 neurons in mediating transsituational stressor outcomes. Uncontrollable (inescapable) stress, but not physically identical controllable (escapable) stress, produced social avoidance and exaggerated fear in male mice. Uncontrollable stress in females led to exploratory avoidance of a novel brightly lit environment. Both controllable and uncontrollable stressors increased VTA VGluT2 neuronal activity, and chemogenetic silencing of VTA VGluT2 neurons prevented the behavioral sequelae of uncontrollable stress in male and female mice. Further, we show that stress activates multiple genetically-distinct subtypes of VTA VGluT2 neurons, especially those that are VGluT2+VGaT+, as well as lateral habenula neurons receiving synaptic input from VTA VGluT2 neurons. Our results provide causal evidence that mice can be used for identifying stressor controllability circuitry and that VTA VGluT2 neurons contribute to transsituational stressor outcomes, such as social avoidance, exaggerated fear, or anxiety-like behavior that are observed within trauma-related disorders.
Collapse
Affiliation(s)
- Dillon J McGovern
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, 80301, CO, US
| | - Annie Ly
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, 80301, CO, US
| | - Koy L Ecton
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, 80301, CO, US
| | - David T Huynh
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, 80301, CO, US
| | - Emily D Prévost
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, 80301, CO, US
| | - Shamira C Gonzalez
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, 80301, CO, US
| | - Connor J McNulty
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, 80301, CO, US
| | - Andrew R Rau
- Department of Biomedical Sciences, Colorado State University, 1617 Campus Delivery, Fort Collins, 80523, CO, US
- Center for Structural and Functional Neuroscience, Division of Biological Sciences, University of Montana, Missoula, 59812, MT, US
| | - Shane T Hentges
- Department of Biomedical Sciences, Colorado State University, 1617 Campus Delivery, Fort Collins, 80523, CO, US
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, 99164, WA, US
| | - Tanya L Daigle
- Allen Institute for Brain Science, 615 Westlake. Avenue North, Seattle, 98109, WA, US
| | - Bosiljka Tasic
- Allen Institute for Brain Science, 615 Westlake. Avenue North, Seattle, 98109, WA, US
| | - Michael V Baratta
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, 80301, CO, US.
| | - David H Root
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, 80301, CO, US.
| |
Collapse
|
9
|
Medeiros D, Ayala-Baylon K, Egido-Betancourt H, Miller E, Chapleau C, Robinson H, Phillips ML, Yang T, Longo FM, Li W, Pozzo-Miller L. A small-molecule TrkB ligand improves dendritic spine phenotypes and atypical behaviors in female Rett syndrome mice. Dis Model Mech 2024; 17:dmm050612. [PMID: 38785269 PMCID: PMC11139040 DOI: 10.1242/dmm.050612] [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: 11/17/2023] [Accepted: 03/06/2024] [Indexed: 05/25/2024] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in MECP2, which encodes methyl-CpG-binding protein 2, a transcriptional regulator of many genes, including brain-derived neurotrophic factor (BDNF). BDNF levels are lower in multiple brain regions of Mecp2-deficient mice, and experimentally increasing BDNF levels improve atypical phenotypes in Mecp2 mutant mice. Due to the low blood-brain barrier permeability of BDNF itself, we tested the effects of LM22A-4, a brain-penetrant, small-molecule ligand of the BDNF receptor TrkB (encoded by Ntrk2), on dendritic spine density and form in hippocampal pyramidal neurons and on behavioral phenotypes in female Mecp2 heterozygous (HET) mice. A 4-week systemic treatment of Mecp2 HET mice with LM22A-4 restored spine volume in MeCP2-expressing neurons to wild-type (WT) levels, whereas spine volume in MeCP2-lacking neurons remained comparable to that in neurons from female WT mice. Female Mecp2 HET mice engaged in aggressive behaviors more than WT mice, the levels of which were reduced to WT levels by the 4-week LM22A-4 treatment. These data provide additional support to the potential usefulness of novel therapies not only for RTT but also to other BDNF-related disorders.
Collapse
Affiliation(s)
- Destynie Medeiros
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Karen Ayala-Baylon
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hailey Egido-Betancourt
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Eric Miller
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christopher Chapleau
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Holly Robinson
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Mary L. Phillips
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Tao Yang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Frank M. Longo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wei Li
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lucas Pozzo-Miller
- Department of Neurobiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
10
|
Jabra S, Rietsche M, Muellerleile J, O'Leary A, Slattery DA, Deller T, Fellenz M. Sex- and cycle-dependent changes in spine density and size in hippocampal CA2 neurons. Sci Rep 2024; 14:12252. [PMID: 38806649 PMCID: PMC11133407 DOI: 10.1038/s41598-024-62951-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024] Open
Abstract
Sex hormones affect structural and functional plasticity in the rodent hippocampus. However, hormone levels not only differ between males and females, but also fluctuate across the female estrous cycle. While sex- and cycle-dependent differences in dendritic spine density and morphology have been found in the rodent CA1 region, but not in the CA3 or the dentate gyrus, comparable structural data on CA2, i.e. the hippocampal region involved in social recognition memory, is so far lacking. In this study, we, therefore, used wildtype male and female mice in diestrus or proestrus to analyze spines on dendritic segments from identified CA2 neurons. In basal stratum oriens, we found no differences in spine density, but a significant shift towards larger spine head areas in male mice compared to females. Conversely, in apical stratum radiatum diestrus females had a significantly higher spine density, and females in either cycle stage had a significant shift towards larger spine head areas as compared to males, with diestrus females showing the larger shift. Our results provide further evidence for the sexual dimorphism of hippocampal area CA2, and underscore the importance of considering not only the sex, but also the stage of the estrous cycle when interpreting morphological data.
Collapse
Affiliation(s)
- Sharif Jabra
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Michael Rietsche
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Julia Muellerleile
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Aet O'Leary
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Goethe University Frankfurt, University Hospital, Heinrich-Hoffmann-Straße 10, 60528, Frankfurt am Main, Germany
| | - David A Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Goethe University Frankfurt, University Hospital, Heinrich-Hoffmann-Straße 10, 60528, Frankfurt am Main, Germany
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Meike Fellenz
- Institute of Clinical Neuroanatomy, Neuroscience Center, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany.
| |
Collapse
|
11
|
Tyson HR, Harrison DJ, Higgs MJ, Isles AR, John RM. Deficiency of the paternally-expressed imprinted Peg3 gene in mice has sexually dimorphic consequences for offspring communication and social behaviour. Front Neurosci 2024; 18:1374781. [PMID: 38595977 PMCID: PMC11002209 DOI: 10.3389/fnins.2024.1374781] [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: 01/22/2024] [Accepted: 03/08/2024] [Indexed: 04/11/2024] Open
Abstract
Introduction Imprinted genes are expressed from one parental allele as a consequence of epigenetic processes initiated in the germline. Consequently, their ability to influence phenotype depends on their parent-of-origin. Recent research suggests that the sex of the individual expressing the imprinted gene is also important. We have previously reported that genetically wildtype (WT) dams carrying and caring for pups mutant for PEG3 exhibit anxiety-like behaviours and their mutant pups show a reduction in ultrasonic vocalisation when separated from their mothers. Sex-specificity was not examined. Methods WT female mice were mated with WT, heterozygous Peg3-/+ or homozygous Peg3-/- studs to generate all WT (control), 50:50 mixed or 100% mutant litters, respectively, followed by behavioural assessment of both dams and their pups. Results We reproduced our original finding that WT dams carrying and caring for 100% mutant litters exhibit postpartum anxiety-like symptoms and delayed pup retrieval. Additionally, these WT dams were found to allocate less time to pup-directed care behaviours relative to controls. Male Peg3-deficient pups demonstrated significantly reduced vocalisation with a more subtle communication deficit in females. Postweaning, male mutants exhibited deficits across a number of key social behaviours as did WT males sharing their environment with mutants. Only modest variations in social behaviour were detected in experimental females. Discussion We have experimentally demonstrated that Peg3 deficiency confined to the offspring causes anxiety in mouse mothers and atypical behaviour including deficits in communication in their male offspring. A male-specific reduction in expression PEG3 in the fetally-derived placenta has previously been associated with maternal depression in human pregnancy. Maternal mood disorders such as depression and anxiety are associated with delays in language development and neuroatypical behaviour more common in sons. Peg3 deficiency could drive the association of maternal and offspring behavioural disorders reported in humans.
Collapse
Affiliation(s)
- Hannah R. Tyson
- Biomedicine Division, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - David J. Harrison
- Biomedicine Division, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Mathew J. Higgs
- Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Anthony R. Isles
- Behavioural Genetics Group, MRC Centre for Neuropsychiatric Genetics and Genomics, Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, United Kingdom
| | - Rosalind M. John
- Biomedicine Division, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
12
|
Ågmo A. Androgen receptors and sociosexual behaviors in mammals: The limits of generalization. Neurosci Biobehav Rev 2024; 157:105530. [PMID: 38176634 DOI: 10.1016/j.neubiorev.2023.105530] [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: 10/18/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 01/06/2024]
Abstract
Circulating testosterone is easily aromatized to estradiol and reduced to dihydrotestosterone in target tissues and elsewhere in the body. Thus, the actions of testosterone can be mediated either by the estrogen receptors, the androgen receptor or by simultaneous action at both receptors. To determine the role of androgens acting at the androgen receptor, we need to eliminate actions at the estrogen receptors. Alternatively, actions at the androgen receptor itself can be eliminated. In the present review, I will analyze the specific role of androgen receptors in male and female sexual behavior as well as in aggression. Some comments about androgen receptors and social recognition are also made. It will be shown that there are important differences between species, even between strains within a species, concerning the actions of the androgen receptor on the behaviors mentioned. This fact makes generalizations from one species to another or from one strain to another very risky. The existence of important species differences is often ignored, leading to many misunderstandings and much confusion.
Collapse
Affiliation(s)
- Anders Ågmo
- Department of Psychology, University of Tromsø, Norway.
| |
Collapse
|
13
|
Cum M, Santiago Pérez JA, Wangia E, Lopez N, Wright ES, Iwata RL, Li A, Chambers AR, Padilla-Coreano N. A systematic review and meta-analysis of how social memory is studied. Sci Rep 2024; 14:2221. [PMID: 38278973 PMCID: PMC10817899 DOI: 10.1038/s41598-024-52277-z] [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: 06/20/2023] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
Social recognition is crucial for survival in social species, and necessary for group living, selective reproduction, pair bonding, and dominance hierarchies. Mice and rats are the most commonly used animal models in social memory research, however current paradigms do not account for the complex social dynamics they exhibit in the wild. To assess the range of social memories being studied, we conducted a systematic analysis of neuroscience articles testing the social memory of mice and rats published within the past two decades and analyzed their methods. Our results show that despite these rodent's rich social memory capabilities, the majority of social recognition papers explore short-term memories and short-term familiarity levels with minimal exposure between subject and familiar stimuli-a narrow type of social memory. We have identified several key areas currently understudied or underrepresented: kin relationships, mates, social ranks, sex variabilities, and the effects of aging. Additionally, reporting on social stimulus variables such as housing history, strain, and age, is limited, which may impede reproducibility. Overall, our data highlight large gaps in the diversity of social memories studied and the effects social variables have on social memory mechanisms.
Collapse
Affiliation(s)
- Meghan Cum
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | | | - Erika Wangia
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | - Naeliz Lopez
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | - Elizabeth S Wright
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | - Ryo L Iwata
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | - Albert Li
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | - Amelia R Chambers
- Department of Neuroscience, University of Florida, Gainesville, 32610, USA
| | | |
Collapse
|
14
|
Cum M, Pérez JS, Wangia E, Lopez N, Wright ES, Iwata RL, Li A, Chambers AR, Padilla-Coreano N. Mind the gap: A systematic review and meta-analysis of how social memory is studied. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.20.572606. [PMID: 38187659 PMCID: PMC10769336 DOI: 10.1101/2023.12.20.572606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Social recognition is crucial for survival in social species, and necessary for group living, selective reproduction, pair bonding, and dominance hierarchies. Mice and rats are the most commonly used animal models in social memory research, however current paradigms do not account for the complex social dynamics they exhibit in the wild. To assess the range of social memories being studied, we conducted a systematic analysis of neuroscience articles testing the social memory of mice and rats published within the past two decades and analyzed their methods. Our results show that despite these rodent's rich social memory capabilities, the majority of social recognition papers explore short-term memories and short-term familiarity levels with minimal exposure between subject and familiar stimuli - a narrow type of social memory. We have identified several key areas currently understudied or underrepresented: kin relationships, mates, social ranks, sex variabilities, and the effects of aging. Additionally, reporting on social stimulus variables such as housing history, strain, and age, is limited, which may impede reproducibility. Overall, our data highlight large gaps in the diversity of social memories studied and the effects social variables have on social memory mechanisms.
Collapse
|
15
|
Medeiros D, Ayala-Baylon K, Egido-Betancourt H, Miller E, Chapleau CA, Robinson HA, Phillips ML, Yang T, Longo F, Li W, Pozzo-Miller L. A small-molecule TrkB ligand improves dendritic spine phenotypes and atypical behaviors in female Rett syndrome mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.09.566435. [PMID: 37986936 PMCID: PMC10659425 DOI: 10.1101/2023.11.09.566435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by mutations in methyl-CpG-binding protein-2 (MECP2), encoding a transcriptional regulator of many genes, including brain-derived neurotrophic factor (Bdnf). BDNF mRNA and protein levels are lower in RTT autopsy brains and in multiple brain regions of Mecp2-deficient mice, and experimentally increasing BDNF levels improve atypical phenotypes in Mecp2 mutant mice. Due to the low blood-brain barrier permeability of BDNF itself, we tested the effects of a brain penetrant, small molecule ligand of its TrkB receptors. Applied in vitro, LM22A-4 increased dendritic spine density in pyramidal neurons in cultured hippocampal slices from postnatal day (P) 7 male Mecp2 knockout (KO) mice as much as recombinant BDNF, and both effects were prevented by the TrkB receptor inhibitors K-252a and ANA-12. Consistent with its partial agonist activity, LM22A-4 did not affect spine density in CA1 pyramidal neurons in slice cultures from male wildtype (WT) mice, where typical BDNF levels outcompete its binding to TrkB. To identify neurons of known genotypes in the "mosaic" brain of female Mecp2 heterozygous (HET) mice, we treated 4-6-month-old female MeCP2-GFP WT and HET mice with peripheral injections of LM22A-4 for 4 weeks. Surprisingly, mutant neurons lacking MeCP2-GFP showed dendritic spine volumes comparable to that in WT controls, while MeCP2-GFP-expressing neurons showed larger spines, similar to the phenotype we described in symptomatic male Mecp2 KO mice where all neurons lack MeCP2. Consistent with this non-cell-autonomous mechanism, a 4-week systemic treatment with LM22A-4 had an effect only in MeCP2-GFP-expressing neurons in female Mecp2 HET mice, bringing dendritic spine volumes down to WT control levels, and without affecting spines of MeCP2-GFP-lacking neurons. At the behavioral level, we found that female Mecp2 HET mice engaged in aggressive behaviors significantly more than WT controls, which were reduced to WT levels by a 4-week systemic treatment with LM22A-4. Altogether, these data revealed differences in dendritic spine size and altered behaviors in Mecp2 HET mice, while providing support to the potential usefulness of BDNF-related therapeutic approaches such as the partial TrkB agonist LM22A-4.
Collapse
|
16
|
Mansk LMZ, Jaimes LF, Dias TL, Pereira GS. Social recognition memory differences between mouse strains: On the effects of social isolation, adult neurogenesis, and environmental enrichment. Brain Res 2023; 1819:148535. [PMID: 37595660 DOI: 10.1016/j.brainres.2023.148535] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 07/25/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
Remembering conspecifics is paramount for the establishment and maintenance of groups. Here we asked whether the variability in social behavior caused by different breeding strategies affects social recognition memory (SRM). We tested the hypothesis that the inbred Swiss and the outbred C57BL/6 mice behave differently on SRM. Social memory in C57BL/6 mice endured at least 14 days, while in Swiss mice lasted 24 h but not ten days. We showed previously that an enriched environment enhanced the persistence of SRM in Swiss mice. Here we reproduced this result and added that it also increases the survival of adult-born neurons in the hippocampus. Next, we tested whether prolonged SRM observed in C57BL/6 mice could be changed by diminishing the trial duration or using an interference stimulus after learning. Neither short acquisition time nor interference during consolidation affected it. However, social isolation impaired SRM in C57BL/6 mice, similar to what was previously observed in Swiss mice. Our results demonstrate that SRM expression can vary according to the mouse strain, which shows the importance of considering this variable when choosing the most suitable model to answer specific questions about this memory system. We also demonstrate the suitability of both C57BL/6 and Swiss strains for exploring the impact of environmental conditions and adult neurogenesis on social memory.
Collapse
Affiliation(s)
- Lara M Z Mansk
- Núcleo de Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Laura F Jaimes
- Núcleo de Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thomaz L Dias
- Núcleo de Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Grace S Pereira
- Núcleo de Neurociências, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
17
|
Hung YC, Wu YJ, Chien ME, Lin YT, Tsai CF, Hsu KS. Loss of oxytocin receptors in hilar mossy cells impairs social discrimination. Neurobiol Dis 2023; 187:106311. [PMID: 37769745 DOI: 10.1016/j.nbd.2023.106311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 09/24/2023] [Accepted: 09/25/2023] [Indexed: 10/02/2023] Open
Abstract
Hippocampal oxytocin receptor (OXTR) signaling is crucial for discrimination of social stimuli to guide social recognition, but circuit mechanisms and cell types involved remain incompletely understood. Here, we report a role for OXTR-expressing hilar mossy cells (MCs) of the dentate gyrus in social stimulus discrimination by regulating granule cell (GC) activity. Using a Cre-loxP recombination approach, we found that ablation of Oxtr from MCs impairs discrimination of social, but not object, stimuli in adult male mice. Ablation of MC Oxtr increases spontaneous firing rate of GCs, synaptic excitation to inhibition ratio of MC-to-GC circuit, and GC firing when temporally associated with the lateral perforant path inputs. Using mouse hippocampal slices, we found that bath application of OXTR agonist [Thr4,Gly7]-oxytocin causes membrane depolarization and increases MC firing activity. Optogenetic activation of MC-to-GC circuit ameliorates social discrimination deficit in MC OXTR deficient mice. Together, our results uncover a previously unknown role of MC OXTR signaling for discrimination of social stimuli and delineate a MC-to-GC circuit responsible for social information processing.
Collapse
Affiliation(s)
- Yu-Chieh Hung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yi-Jen Wu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70457, Taiwan; Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70403, Taiwan
| | - Miao-Er Chien
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 70457, Taiwan
| | - Yu-Ting Lin
- Institute of Systems Neuroscience, College of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Cheng-Fang Tsai
- Department of Physical Medicine and Rehabilitation, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi 60002, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
| | - Kuei-Sen Hsu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan; Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan.
| |
Collapse
|
18
|
Granza AE, Amaral IM, Monteiro DG, Salti A, Hofer A, El Rawas R. Social Interaction Is Less Rewarding in Adult Female than in Male Mice. Brain Sci 2023; 13:1445. [PMID: 37891813 PMCID: PMC10605033 DOI: 10.3390/brainsci13101445] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/05/2023] [Accepted: 10/08/2023] [Indexed: 10/29/2023] Open
Abstract
(1) Background: Positive social relationships are essential for mental and physical health. However, not all individuals experience social interaction as a rewarding activity. (2) Methods: Social interaction reward in mice can be assessed by social conditioned place preference (CPP). The aim of this study is to investigate sex-dependent differences in the neurological underpinnings underlying social versus non-social phenotypes, using adult male and female C57BL/6J mice. (3) Results: Adult female mice expressed significantly less social reward than males from the same strain. Accordingly, pairs of male mice spent more time interacting as compared to female pairs. Subsequently, we analyzed neuropeptides previously reported to be important regulators of social behavior such as oxytocin, vasopressin, and orexin, in addition to Ca2+/calmodulin-dependent protein kinase II (αCaMKII), shown to be involved in social reward. Levels of neuropeptides and αCaMKII were comparable between males and females in all investigated regions. Yet, a significant negative correlation was found between endogenous oxytocin expression and social reward in female pairs. (4) Conclusions: Sex differences in the prevalence of many mental health disorders might at least in part be due to sex differences in social reward. Therefore, more research is needed to unravel the candidate(s) underlying this behavioral difference.
Collapse
Affiliation(s)
- Anna E. Granza
- Division of Psychiatry I, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Inês M. Amaral
- Division of Psychiatry I, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Diogo G. Monteiro
- Division of Psychiatry I, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Ahmad Salti
- University Clinic of Ophthalmology and Optometry, Kepler University Hospital, Johannes Kepler University Linz, Krankenhausstrasse 5, 4020 Linz, Austria
| | - Alex Hofer
- Division of Psychiatry I, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Rana El Rawas
- Division of Psychiatry I, Department of Psychiatry, Psychotherapy, Psychosomatics and Medical Psychology, Medical University Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
19
|
Guo M, Sun L. From rodents to humans: Rodent behavioral paradigms for social behavioral disorders. Brain Circ 2023; 9:154-161. [PMID: 38020957 PMCID: PMC10679632 DOI: 10.4103/bc.bc_48_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/06/2023] [Accepted: 08/09/2023] [Indexed: 12/01/2023] Open
Abstract
Social cognition guides social behavior. Subjects with proper social cognition should be able to: (1) have reasonable social motivation, (2) recognize other people and infer their intentions, and (3) weigh social hierarchies and other values. The choice of appropriate behavioral paradigms enables the use of rodents to study social behavior disorders in humans, thus enabling research to go deeper into neural mechanisms. This paper reviews commonly used rodent behavioral paradigms in studies of social behavior disorders. We focused specifically on sorting out ways to transfer the study of human social behavior to rodents through behavioral paradigms.
Collapse
Affiliation(s)
- Mingyue Guo
- Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Capital Medical University, Beijing, China
| | - Le Sun
- Collaborative Innovation Center for Brain Disorders, Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Capital Medical University, Beijing, China
| |
Collapse
|
20
|
Yoest KE, Henry MG, Velisek HA, Veenema AH. Development of social recognition ability in female rats: Effect of pubertal ovarian hormones. Horm Behav 2023; 151:105347. [PMID: 36966657 DOI: 10.1016/j.yhbeh.2023.105347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 04/28/2023]
Abstract
The ability to recognize previously encountered conspecifics is crucial for social interaction. This social recognition ability is well characterized in adult rodents of both sexes but remains largely unexplored in juveniles. Using the social discrimination test of social recognition with short intervals (30 min and 1 h), we first found that juvenile female rats do not display a difference in investigation directed toward a novel vs. familiar stimulus rat. Using the social discrimination test with a 30-minute interval, we then showed that social recognition is established by the time of adolescence in female rats. Based on these findings, we hypothesized that social recognition is dependent on the initiation of ovarian hormone release during puberty. To test this, we ovariectomized females prior to puberty and found that prepubertal ovariectomy prevented the development of social recognition ability in adulthood. Administration of estradiol benzoate, 48 h prior to testing, to juvenile females or prepubertally ovariectomized adult females did not restore social recognition, suggesting that ovarian hormones organize the neural circuitry regulating this behavior during adolescence. These findings provide the first evidence of an effect of pubertal development on social recognition ability in female rats and highlight the importance of considering sex and age when interpreting results from behavioral paradigms initially designed for use in adult males.
Collapse
Affiliation(s)
- Katie E Yoest
- Department of Psychology, Michigan State University, East Lansing, MI, United States of America
| | - Morgen G Henry
- Department of Psychology, Michigan State University, East Lansing, MI, United States of America
| | - Haley A Velisek
- Department of Psychology, Michigan State University, East Lansing, MI, United States of America
| | - Alexa H Veenema
- Department of Psychology, Michigan State University, East Lansing, MI, United States of America; Neuroscience Program, Michigan State University, East Lansing, MI, United States of America.
| |
Collapse
|
21
|
Suarez LM, Diaz-Del Cerro E, Felix J, Gonzalez-Sanchez M, Ceprian N, Guerra-Perez N, G Novelle M, Martinez de Toda I, De la Fuente M. Sex differences in neuroimmunoendocrine communication. Involvement on longevity. Mech Ageing Dev 2023; 211:111798. [PMID: 36907251 DOI: 10.1016/j.mad.2023.111798] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/07/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023]
Abstract
Endocrine, nervous, and immune systems work coordinately to maintain the global homeostasis of the organism. They show sex differences in their functions that, in turn, contribute to sex differences beyond reproductive function. Females display a better control of the energetic metabolism and improved neuroprotection and have more antioxidant defenses and a better inflammatory status than males, which is associated with a more robust immune response than that of males. These differences are present from the early stages of life, being more relevant in adulthood and influencing the aging trajectory in each sex and may contribute to the different life lifespan between sexes.
Collapse
Affiliation(s)
- Luz M Suarez
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain.
| | - Estefania Diaz-Del Cerro
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain
| | - Judith Felix
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain
| | - Monica Gonzalez-Sanchez
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain
| | - Noemi Ceprian
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain
| | - Natalia Guerra-Perez
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain
| | - Marta G Novelle
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain
| | - Irene Martinez de Toda
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain
| | - Monica De la Fuente
- Department of Genetics, Physiology, and Microbiology (Unit of Animal Physiology), Faculty of Biology, Complutense University, Madrid, Spain; Institute of Investigation Hospital 12 Octubre (imas12), Madrid, Spain.
| |
Collapse
|
22
|
Maletta T, Palummieri M, Correa J, Holahan MR. Preadolescent exposure to a sexually mature, unrelated male rat reduces postadolescent social recognition memory and CA2 c-Fos labeling. Front Behav Neurosci 2023; 17:1104866. [PMID: 36778132 PMCID: PMC9908592 DOI: 10.3389/fnbeh.2023.1104866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/13/2023] [Indexed: 01/27/2023] Open
Abstract
Introduction Social memory involves social recognition: the ability to discriminate between two or more conspecifics when one has been previously encountered. The CA2 region of the hippocampus has been implicated in social memory, as lesions and dysfunction to this area lead to social memory impairments. A variety of psychogenic manipulations during postnatal sensitive developmental periods are associated with social memory impairments later in life. Methods In this study, we exposed preadolescent rats to a sexually, mature unrelated male and examined whether this was associated with changes in postadolescent social memory and c-Fos labeling in the CA2 region. Male and female Long-Evans rats were exposed to a male, adult rat on postnatal days 19-21 (P19-21). Social memory was measured during the postadolescent period and defined as increased interactions towards a novel age-matched rat in contrast to a previously-encountered age-matched rat. After the test, rats were euthanized and brain tissue was then collected to quantify c-Fos labeling within the CA2 region. Results Compared to home cage controls and controls not exposed to the adult male, male and female rats exposed to the unrelated adult during preadolescence were unable to discriminate between a novel and previously encountered conspecific during the postadolescent test showing social memory deficits. The groups that showed social recognition deficits also had significantly fewer c-Fos-positive cells within the CA2 region compared to the control groups. Discussion These findings indicate that threatening psychogenic encounters during preadolescence can have detrimental long-term effects on social memory potentially via disrupted activity in the CA2 hippocampal region.
Collapse
Affiliation(s)
- Teresa Maletta
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | | | - Jeff Correa
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | | |
Collapse
|
23
|
Rigney N, de Vries GJ, Petrulis A. Modulation of social behavior by distinct vasopressin sources. Front Endocrinol (Lausanne) 2023; 14:1127792. [PMID: 36860367 PMCID: PMC9968743 DOI: 10.3389/fendo.2023.1127792] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 01/30/2023] [Indexed: 02/15/2023] Open
Abstract
The neuropeptide arginine-vasopressin (AVP) is well known for its peripheral effects on blood pressure and antidiuresis. However, AVP also modulates various social and anxiety-related behaviors by its actions in the brain, often sex-specifically, with effects typically being stronger in males than in females. AVP in the nervous system originates from several distinct sources which are, in turn, regulated by different inputs and regulatory factors. Based on both direct and indirect evidence, we can begin to define the specific role of AVP cell populations in social behavior, such as, social recognition, affiliation, pair bonding, parental behavior, mate competition, aggression, and social stress. Sex differences in function may be apparent in both sexually-dimorphic structures as well as ones without prominent structural differences within the hypothalamus. The understanding of how AVP systems are organized and function may ultimately lead to better therapeutic interventions for psychiatric disorders characterized by social deficits.
Collapse
Affiliation(s)
- Nicole Rigney
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | | | | |
Collapse
|
24
|
Rodriguez LA, Kim SH, Page SC, Nguyen CV, Pattie EA, Hallock HL, Valerino J, Maynard KR, Jaffe AE, Martinowich K. The basolateral amygdala to lateral septum circuit is critical for regulating social novelty in mice. Neuropsychopharmacology 2023; 48:529-539. [PMID: 36369482 PMCID: PMC9852457 DOI: 10.1038/s41386-022-01487-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 10/07/2022] [Accepted: 10/24/2022] [Indexed: 11/13/2022]
Abstract
The lateral septum (LS) is a basal forebrain GABAergic region that is implicated in social novelty. However, the neural circuits and cell signaling pathways that converge on the LS to mediate social behaviors aren't well understood. Multiple lines of evidence suggest that signaling of brain-derived neurotrophic factor (BDNF) through its receptor TrkB plays important roles in social behavior. BDNF is not locally produced in LS, but we demonstrate that nearly all LS GABAergic neurons express TrkB. Local TrkB knock-down in LS neurons decreased social novelty recognition and reduced recruitment of neural activity in LS neurons in response to social novelty. Since BDNF is not synthesized in LS, we investigated which inputs to LS could serve as potential BDNF sources for controlling social novelty recognition. We demonstrate that selectively ablating inputs to LS from the basolateral amygdala (BLA), but not from ventral CA1 (vCA1), impairs social novelty recognition. Moreover, depleting BDNF selectively in BLA-LS projection neurons phenocopied the decrease in social novelty recognition caused by either local LS TrkB knockdown or ablation of BLA-LS inputs. These data support the hypothesis that BLA-LS projection neurons serve as a critical source of BDNF for activating TrkB signaling in LS neurons to control social novelty recognition.
Collapse
Affiliation(s)
- Lionel A Rodriguez
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Sun-Hong Kim
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Stephanie C Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Claudia V Nguyen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Elizabeth A Pattie
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Henry L Hallock
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Jessica Valerino
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Andrew E Jaffe
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Genetic Medicine, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Keri Martinowich
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, USA.
| |
Collapse
|
25
|
Pinizzotto CC, Patwardhan A, Aldarondo D, Kritzer MF. Task-specific effects of biological sex and sex hormones on object recognition memories in a 6-hydroxydopamine-lesion model of Parkinson's disease in adult male and female rats. Horm Behav 2022; 144:105206. [PMID: 35653829 DOI: 10.1016/j.yhbeh.2022.105206] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/19/2022] [Accepted: 05/21/2022] [Indexed: 01/13/2023]
Abstract
Many patients with Parkinson's disease (PD) experience cognitive or memory impairments with few therapeutic options available to mitigate them. This has fueled interest in determining how factors including sex and sex hormones modulate higher order function in this disease. The objective of this study was to use the Novel Object Recognition (NOR) and Object-in-Place (OiP) paradigms to compare the effects of a bilateral neostriatal 6-hydroxydopamine (6-OHDA) lesion model of PD in gonadally intact male and female rats, in orchidectomized male rats and in orchidectomized males supplemented with 17β-estradiol or testosterone propionate on measures of recognition memory similar to those at risk in PD. These studies showed that 6-ODHA lesions impaired discrimination in both tasks in males but not females. Further, 6-OHDA lesions disrupted NOR performance similarly in all males regardless of whether they were gonadally intact, orchidectomized or hormone-supplemented. In contrast, OiP performance was disrupted in males that were orchidectomized or 6-OHDA-lesioned but was spared in orchidectomized and orchidectomized, 6-OHDA lesioned males supplemented with 17β-estradiol. The distinct effects that sex and/or sex hormones have on 6-OHDA lesion-induced NOR vs. OiP deficits identified here also differ from corresponding impacts recently described for 6-OHDA lesion-induced deficits in spatial working memory and episodic memory. Together, the collective data provide strong evidence for effects of sex and sex hormones on cognition and memory in PD as being behavioral task and behavioral domain specific. This specificity could explain why a cohesive clinical picture of endocrine impacts on higher order function in PD has remained elusive.
Collapse
Affiliation(s)
- Claudia C Pinizzotto
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York 11794, USA.
| | - Aishwarya Patwardhan
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York 11794, USA.
| | - Daniel Aldarondo
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York 11794, USA.
| | - Mary F Kritzer
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, New York 11794, USA.
| |
Collapse
|
26
|
Tsai TC, Fang YS, Hung YC, Hung LC, Hsu KS. A dorsal CA2 to ventral CA1 circuit contributes to oxytocinergic modulation of long-term social recognition memory. J Biomed Sci 2022; 29:50. [PMID: 35811321 PMCID: PMC9272559 DOI: 10.1186/s12929-022-00834-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/06/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Social recognition memory (SRM) is the ability to distinguish familiar from novel conspecifics and is crucial for survival and reproductive success across social species. We previously reported that oxytocin (OXT) receptor (OXTR) signaling in the CA2/CA3a of dorsal hippocampus is essential to promote the persistence of long-term SRM, yet how the endogenous OXT system influences CA2 outputs to regulate long-term SRM formation remains unclear. METHODS To achieve a selective deletion of CA2 OXTRs, we crossed Amigo2-Cre mice with Oxtr-floxed mice to generate CA2-specific Oxtr conditional knockout (Oxtr-/-) mice. A three-chamber paradigm test was used for studying SRM in mice. Chemogenetic and optogenetic targeting strategies were employed to manipulate neuronal activity. RESULTS We show that selective ablation of Oxtr in the CA2 suffices to impair the persistence of long-term SRM but has no effect on sociability and social novelty preference in the three-chamber paradigm test. We find that cell-type specific activation of OXT neurons within the hypothalamic paraventricular nucleus enhances long-term SRM and this enhancement is blocked by local application of OXTR antagonist L-368,899 into dorsal hippocampal CA2 (dCA2) region. In addition, chemogenetic neuronal silencing in dCA2 demonstrated that neuronal activity is essential for forming long-term SRM. Moreover, chemogenetic terminal-specific inactivation reveals a crucial role for dCA2 outputs to ventral CA1 (vCA1), but not dorsal lateral septum, in long-term SRM. Finally, targeted activation of the dCA2-to-vCA1 circuit effectively ameliorates long-term SRM deficit observed in Oxtr-/- mice. CONCLUSIONS These findings highlight the importance of hippocampal CA2 OXTR signaling in governing the persistence of long-term SRM and identify a hippocampal circuit linking dCA2 to vCA1 necessary for controlling long-term SRM formation.
Collapse
Affiliation(s)
- Tsung-Chih Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Yi-Syuan Fang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No. 1, University Rd., Tainan, 70101, Taiwan
| | - Yu-Chieh Hung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Ling-Chien Hung
- Division of Neurology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, 60002, Taiwan.
| | - Kuei-Sen Hsu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan.
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No. 1, University Rd., Tainan, 70101, Taiwan.
| |
Collapse
|
27
|
High Fructose and High Fat Diet Impair Different Types of Memory through Oxidative Stress in a Sex- and Hormone-Dependent Manner. Metabolites 2022; 12:metabo12040341. [PMID: 35448528 PMCID: PMC9024673 DOI: 10.3390/metabo12040341] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 02/05/2023] Open
Abstract
Metabolic syndrome (MetS) contributes to the spread of cardiovascular diseases, diabetes mellitus type 2, and neurodegenerative diseases. Evaluation of sex- and hormone-dependent changes in body weight, blood pressure, blood lipids, oxidative stress markers, and alterations in different types of memory in Sprague–Dawley rats fed with a high fat and high fructose (HFHF) diet were evaluated. After 12 weeks of feeding the male and female rats with HFHF, body weight gain, increase in blood pressure, and generation of dyslipidemia compared to the animals fed with chow diet were observed. Regarding memory, it was noted that gonadectomy reverted the effects of HFHF in the 24 h novel object recognition task and in spatial learning/memory analyzed through Morris water maze, males being more affected than females. Nevertheless, gonadectomy did not revert long-term memory impairment in the passive avoidance task induced by HFHF nor in male or female rats. On the other hand, sex-hormone–diet interaction was observed in the plasma concentration of malondialdehyde and nitric oxide. These results suggest that the changes observed in the memory and learning of MetS animals are sex- and hormone-dependent and correlate to an increase in oxidative stress.
Collapse
|
28
|
Yaw AM, Glass JD, Prosser RA, Caldwell HK. Paternal Cocaine in Mice Alters Social Behavior and Brain Oxytocin Receptor Density in First Generation Offspring. Neuroscience 2022; 485:65-77. [PMID: 35063583 PMCID: PMC8866213 DOI: 10.1016/j.neuroscience.2022.01.010] [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] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 12/27/2022]
Abstract
It is well established that the damaging effects of drugs of abuse, such as cocaine, can extend beyond the user to their offspring. While most preclinical models of the generational effects of cocaine abuse have focused on maternal effects, we, and others, report distinct effects on offspring sired by fathers treated with cocaine prior to breeding. However, little is known about the effects of paternal cocaine use on first generation (F1) offspring's social behaviors. Here, we expand upon our model of oral self-administered paternal cocaine use to address the idea that paternal cocaine alters first generation offspring social behaviors through modulation of the oxytocin system. F1 cocaine-sired males displayed unaltered social recognition vs. non-cocaine sired controls but showed increased investigation times that were not related to altered olfaction. Paternal cocaine did not alter F1 male-aggression behavior or depression-like behaviors, but cocaine-sired males did display decreased anxiety-like behaviors. Female F1 behavior was similarly examined, but there were no effects of paternal cocaine. Cocaine-sired male mice also exhibited localized oxytocin receptor expression differences vs. controls in several brain regions regulating social behavior. These results provide evidence for effects of paternal cocaine exposure on social behaviors in male offspring with associated alterations in central oxytocin transmission.
Collapse
Affiliation(s)
- Alexandra M Yaw
- School of Biomedical Sciences, Kent State Univ., Kent, OH 44242, United States; Laboratory of Neuroendocrinology and Behavior, Department of Biological Sciences, Brain Health Research Institute, Kent State University, Kent, OH, United States
| | - J David Glass
- School of Biomedical Sciences, Kent State Univ., Kent, OH 44242, United States
| | - Rebecca A Prosser
- Department of Biochemistry & Cellular and Molecular Biology, and the NeuroNET Research Center, Univ. of Tennessee, Knoxville, TN 37996, United States
| | - Heather K Caldwell
- School of Biomedical Sciences, Kent State Univ., Kent, OH 44242, United States; Laboratory of Neuroendocrinology and Behavior, Department of Biological Sciences, Brain Health Research Institute, Kent State University, Kent, OH, United States.
| |
Collapse
|
29
|
Lopez-Rojas J, de Solis CA, Leroy F, Kandel ER, Siegelbaum SA. A direct lateral entorhinal cortex to hippocampal CA2 circuit conveys social information required for social memory. Neuron 2022; 110:1559-1572.e4. [PMID: 35180391 PMCID: PMC9081137 DOI: 10.1016/j.neuron.2022.01.028] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/21/2021] [Accepted: 01/24/2022] [Indexed: 11/18/2022]
Abstract
The hippocampus is essential for different forms of declarative memory, including social memory, the ability to recognize and remember a conspecific. Although recent studies identify the importance of the dorsal CA2 region of the hippocampus in social memory storage, little is known about its sources of social information. Because CA2, like other hippocampal regions, receives its major source of spatial and non-spatial information from the medial and lateral subdivisions of entorhinal cortex (MEC and LEC), respectively, we investigated the importance of these inputs for social memory. Whereas MEC inputs to CA2 are dispensable, the direct inputs to CA2 from LEC are both selectively activated during social exploration and required for social memory. This selective behavioral role of LEC is reflected in the stronger excitatory drive it provides to CA2 compared with MEC. Thus, a direct LEC → CA2 circuit is tuned to convey social information that is critical for social memory.
Collapse
Affiliation(s)
- Jeffrey Lopez-Rojas
- Department of Neuroscience, The Kavli Institute for Brain Science, Mortimer B. Zuckerman Mind Brain Behavior Institute, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA.
| | - Christopher A de Solis
- Department of Neuroscience, The Kavli Institute for Brain Science, Mortimer B. Zuckerman Mind Brain Behavior Institute, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA
| | - Felix Leroy
- Department of Neuroscience, The Kavli Institute for Brain Science, Mortimer B. Zuckerman Mind Brain Behavior Institute, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA; Instituto de Neurociencias CSIC-UMH, San Juan de Alicante, Spain
| | - Eric R Kandel
- Department of Neuroscience, The Kavli Institute for Brain Science, Mortimer B. Zuckerman Mind Brain Behavior Institute, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA; Howard Hughes Medical Institute, Columbia University, New York, NY, USA
| | - Steven A Siegelbaum
- Department of Neuroscience, The Kavli Institute for Brain Science, Mortimer B. Zuckerman Mind Brain Behavior Institute, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
30
|
Gillette R, Dias M, Reilly MP, Thompson LM, Castillo NJ, Vasquez EL, Crews D, Gore AC. Two Hits of EDCs Three Generations Apart: Effects on Social Behaviors in Rats, and Analysis by Machine Learning. TOXICS 2022; 10:toxics10010030. [PMID: 35051072 PMCID: PMC8779176 DOI: 10.3390/toxics10010030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/12/2021] [Accepted: 01/07/2022] [Indexed: 02/07/2023]
Abstract
All individuals are directly exposed to extant environmental endocrine-disrupting chemicals (EDCs), and indirectly exposed through transgenerational inheritance from our ancestors. Although direct and ancestral exposures can each lead to deficits in behaviors, their interactions are not known. Here we focused on social behaviors based on evidence of their vulnerability to direct or ancestral exposures, together with their importance in reproduction and survival of a species. Using a novel "two hits, three generations apart" experimental rat model, we investigated interactions of two classes of EDCs across six generations. PCBs (a weakly estrogenic mixture Aroclor 1221, 1 mg/kg), Vinclozolin (antiandrogenic, 1 mg/kg) or vehicle (6% DMSO in sesame oil) were administered to pregnant rat dams (F0) to directly expose the F1 generation, with subsequent breeding through paternal or maternal lines. A second EDC hit was given to F3 dams, thereby exposing the F4 generation, with breeding through the F6 generation. Approximately 1200 male and female rats from F1, F3, F4 and F6 generations were run through tests of sociability and social novelty as indices of social preference. We leveraged machine learning using DeepLabCut to analyze nuanced social behaviors such as nose touching with accuracy similar to a human scorer. Surprisingly, social behaviors were affected in ancestrally exposed but not directly exposed individuals, particularly females from a paternally exposed breeding lineage. Effects varied by EDC: Vinclozolin affected aspects of behavior in the F3 generation while PCBs affected both the F3 and F6 generations. Taken together, our data suggest that specific aspects of behavior are particularly vulnerable to heritable ancestral exposure of EDC contamination, that there are sex differences, and that lineage is a key factor in transgenerational outcomes.
Collapse
Affiliation(s)
- Ross Gillette
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (R.G.); (M.D.); (M.P.R.); (L.M.T.); (N.J.C.); (E.L.V.)
| | - Michelle Dias
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (R.G.); (M.D.); (M.P.R.); (L.M.T.); (N.J.C.); (E.L.V.)
| | - Michael P. Reilly
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (R.G.); (M.D.); (M.P.R.); (L.M.T.); (N.J.C.); (E.L.V.)
| | - Lindsay M. Thompson
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (R.G.); (M.D.); (M.P.R.); (L.M.T.); (N.J.C.); (E.L.V.)
| | - Norma J. Castillo
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (R.G.); (M.D.); (M.P.R.); (L.M.T.); (N.J.C.); (E.L.V.)
| | - Erin L. Vasquez
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (R.G.); (M.D.); (M.P.R.); (L.M.T.); (N.J.C.); (E.L.V.)
| | - David Crews
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, USA;
| | - Andrea C. Gore
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; (R.G.); (M.D.); (M.P.R.); (L.M.T.); (N.J.C.); (E.L.V.)
- Correspondence:
| |
Collapse
|
31
|
Endogenous Estrogen Influences Predator Odor-Induced Impairment of Cognitive and Social Behaviors in Aromatase Gene Deficiency Mice. Behav Neurol 2021; 2021:5346507. [PMID: 34594430 PMCID: PMC8478571 DOI: 10.1155/2021/5346507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/28/2021] [Indexed: 11/17/2022] Open
Abstract
Epidemiological studies have suggested that traumatic stress increases vulnerability to various mental disorders, such as dementia and psychiatric disorders. While women are more vulnerable than men to depression and anxiety, it is unclear whether endogenous estrogens are responsible for the underlying sex-specific mechanisms. In this study, the aromatase gene heterozygous (Ar+/-) mice were used as an endogenous estrogen deficiency model and age- and sex-matched wild type mice (WT) as controls to study the predator odor 2,3,5-trimethyl-3-thiazoline- (TMT-) induced short- and long-term cognitive and social behavior impairments. In addition, the changes in brain regional neurotransmitters and their associations with TMT-induced changes in behaviors were further investigated in these animals. Our results showed TMT induced immediate fear response in both Ar+/- and WT mice regardless of sexes. TMT induced an acute impairment of novel object recognition memory and long-term social behavior impairment in WT mice, particularly in females, while Ar+/- mice showed impaired novel object recognition in both sexes and TMT-elevated social behaviors, particularly in males. TMT failed to induce changes in the prepulse inhibition (PPI) test in both groups. TMT resulted in a slight increase of DOPAC/DA ratio in the cortex and a significant elevation of this ratio in the striatum of WT mice. In addition, the ratio of HIAA/5-HT was significantly elevated in the cortex of TMT-treated WT mice, which was not found in TMT-treated Ar+/- mice. Taken together, our results indicate that TMT exposure can cause cognitive and social behavior impairments as well as change catecholamine metabolism in WT mice, and endogenous estrogen deficiency might desensitize the behavioral and neurochemical responses to TMT in Ar+/- mice.
Collapse
|
32
|
Sex differences in offspring discrimination in the biparental California mouse (Peromyscus californicus). J ETHOL 2021. [DOI: 10.1007/s10164-021-00705-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
33
|
Morin A, Van de Beeck L, Person E, Plamondon H. Adult Male Rats Show Resilience to Adolescent Bisphenol A Effects on Hormonal and Behavioral Responses While Co-Exposure With Hop Extracts Supports Synergistic Actions. FRONTIERS IN TOXICOLOGY 2021; 3:639820. [PMID: 35295120 PMCID: PMC8915799 DOI: 10.3389/ftox.2021.639820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/25/2021] [Indexed: 11/13/2022] Open
Abstract
The adolescence period, marked by sexual and brain maturation, has shown sensitivity to various environmental disruptors. Exposure to the xenoestrogen bisphenol A (BPA) is known to alter physiological and behavioral responses although its role at this critical period remains largely unknown. Recent research further suggests biochemical and genomic effects of BPA to be mitigated by various natural compounds, while effects on behavior have not been examined. This study aimed to characterize (1) the effects of dietary BPA during adolescence on endogenous corticosterone (CORT) secretion, emotional behavior, and testosterone (T) in adulthood, and (2) the impact of combined exposure to BPA with hop extracts (Hop), a phytoestrogen with anxiolytic properties. To do so, four groups of male Wistar rats [postnatal day (PND) 28] were administered corn oil (control), BPA (40 mg/kg), hops (40 mg/kg), or BPA-hops by oral gavage for 21 days (PND 28–48). Blood droplets collected on PND 28, 48, and 71 served to measure CORT and T changes. As adults, rats were tested in the elevated plus maze (EPM), the social interaction test, and the forced swim test. Our findings demonstrated elevated anxiety and a trend toward depressive-like behaviors in BPA- compared to hops-exposed rats. However, BPA intake had no impact on basal CORT levels, or adulthood T secretion and sociability. Of note, BPA's anxiogenic effect manifested through decreased EPM open arm entries was abolished by hops co-supplementation. Together, our observations suggest the adolescence period to be less sensitive to deleterious effects of BPA than what has been reported upon gestational and perinatal exposure.
Collapse
|
34
|
Adu-Nti F, Gao X, Wu JM, Li J, Iqbal J, Ahmad R, Ma XM. Osthole Ameliorates Estrogen Deficiency-Induced Cognitive Impairment in Female Mice. Front Pharmacol 2021; 12:641909. [PMID: 34025413 PMCID: PMC8134730 DOI: 10.3389/fphar.2021.641909] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/04/2021] [Indexed: 12/24/2022] Open
Abstract
Loss of endogenous estrogen and dysregulation of the estrogen receptor signaling pathways are associated with an increase in risk for cognitive deficit and depression in women after menopause. Estrogen therapy for menopause increases the risk of breast and ovarian cancers, and stroke. Therefore, it is critical to find an alternate treatment for menopausal women. Osthole (OST), a coumarin, has been reported to have neuroprotective effects. This study examined whether OST improves ovariectomy (OVX)-induced cognitive impairment, and alleviates anxiety- and depression-like behaviors induced by OVX in mice. Adult female C57BL/6J mice were ovariectomized and then treated with OST at a dose of 30 mg/kg for 14 days. At the end of the treatment period, behavioral tests were used to evaluate spatial learning and memory, recognition memory, anxiety- and depression-like behaviors. A cohort of the mice were sacrificed after 14 days of OST treatment and their hippocampi were collected for measurement of the proteins of interest using western blot. OVX-induced alteration in the levels of proteins was accompanied by cognitive deficit, anxiety- and depression-like behaviors. OST treatment improved cognitive deficit, alleviated anxiety- and depression-like behaviors induced by OVX, and reversed OVX-induced alterations in the levels of synaptic proteins and ERα, BDNF, TrKB, p-CREB, p-Akt and Rac1 in the hippocampus. Therefore, reversal of OVX-induced decrease in the levels of hippocampal proteins by OST might contribute to the effects of OST on improving cognitive deficit and alleviating anxiety- and depression-like behaviors induced by OVX.
Collapse
Affiliation(s)
- Frank Adu-Nti
- Department of Neuroscience, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xu Gao
- Department of Neuroscience, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Jia-Min Wu
- Department of Neuroscience, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Jing Li
- Department of Neuroscience, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Javed Iqbal
- Department of Neuroscience, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Riaz Ahmad
- Department of Neuroscience, Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Chinese Crude Drugs in Northwest China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Xin-Ming Ma
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, United States
| |
Collapse
|
35
|
Rigney N, Whylings J, de Vries GJ, Petrulis A. Sex Differences in the Control of Social Investigation and Anxiety by Vasopressin Cells of the Paraventricular Nucleus of the Hypothalamus. Neuroendocrinology 2021; 111:521-535. [PMID: 32541145 PMCID: PMC7736187 DOI: 10.1159/000509421] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/15/2020] [Indexed: 12/11/2022]
Abstract
The neuropeptide arginine-vasopressin (AVP) has long been implicated in the regulation of social behavior and communication in diverse taxa, but the source of AVP release relevant for behavior has not been precisely determined. Potential sources include hypothalamic cell populations such as the paraventricular (PVN), supraoptic, and suprachiasmatic nuclei, as well as extrahypothalamic cell groups in the extended amygdala. To address if AVP-expressing cells in the PVN are important for mouse social communication, we deleted PVN AVP-expressing cells using viral-mediated delivery of Cre-dependent caspase-9 cell death construct into the PVN of AVP-Cre-positive mice (expressing Cre-recombinase under the control of the AVP promoter) or AVP-Cre-negative littermate controls, and assessed their levels of social investigation, social communication, anxiety, sex behavior, and aggressive behavior. We found that these lesions increased social investigation in females, but not in males. However, in males but not in females, these lesions increased non-social anxiety-related behaviors in the elevated-plus maze. These results therefore point at differential involvement of PVN AVP-expressing cells in the context of social and emotional behavior in the two sexes, which may contribute to sex differences in social communication and anxiety disorders.
Collapse
Affiliation(s)
- Nicole Rigney
- Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA,
| | - Jack Whylings
- Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Geert J de Vries
- Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| | - Aras Petrulis
- Center for Behavioral Neuroscience, Neuroscience Institute, Georgia State University, Atlanta, Georgia, USA
| |
Collapse
|
36
|
Adult-Born Neurons in the Hippocampus Are Essential for Social Memory Maintenance. eNeuro 2020; 7:ENEURO.0182-20.2020. [PMID: 33060182 PMCID: PMC7768285 DOI: 10.1523/eneuro.0182-20.2020] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/18/2020] [Accepted: 09/30/2020] [Indexed: 01/02/2023] Open
Abstract
Throughout adulthood, the dentate gyrus continues to produce new granule cells, which integrate into the hippocampal circuitry. New neurons have been linked to several known functions of the hippocampus, including learning and memory, anxiety and stress regulation, and social behavior. We explored whether transgenic reduction of adult-born neurons in mice would impair social memory and the formation of social dominance hierarchies. We used a conditional transgenic mouse strain [thymidine kinase (TK) mice] that selectively reduces adult neurogenesis by treatment with the antiviral drug valganciclovir (VGCV). TK mice treated with VGCV were unable to recognize conspecifics as familiar 24 h after initial exposure. We then explored whether reducing new neurons completely impaired their ability to acquire or retrieve a social memory and found that TK mice treated with VGCV were able to perform at control levels when the time between exposure (acquisition) and reexposure (retrieval) was brief. We next explored whether adult-born neurons are involved in dominance hierarchy formation by analyzing their home cage behavior as well as their performance in the tube test, a social hierarchy test, and did not find any consistent alterations in behavior between control and TK mice treated with VGCV. These data suggest that adult neurogenesis is essential for social memory maintenance, but not for acquisition nor retrieval over a short time frame, with no effect on social dominance hierarchy. Future work is needed to explore whether the influence of new neurons on social memory is mediated through connections with the CA2, an area involved in social recognition.
Collapse
|
37
|
Scheggi S, Guzzi F, Braccagni G, De Montis MG, Parenti M, Gambarana C. Targeting PPARα in the rat valproic acid model of autism: focus on social motivational impairment and sex-related differences. Mol Autism 2020; 11:62. [PMID: 32718349 PMCID: PMC7385875 DOI: 10.1186/s13229-020-00358-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 06/16/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The social motivational theory of autism spectrum disorder (ASD) focuses on social anhedonia as key causal feature of the impaired peer relationships that characterize ASD patients. ASD prevalence is higher in boys, but increasing evidence suggests underdiagnosis and undertreatment in girls. We showed that stress-induced motivational anhedonia is relieved by repeated treatment with fenofibrate (FBR), a peroxisome proliferator-activated receptor α (PPARα) agonist. Here, we used the valproic acid (VPA) model of ASD in rats to examine male and female phenotypes and assess whether FBR administration from weaning to young adulthood relieved social impairments. METHODS Male and female rats exposed to saline or VPA at gestational day 12.5 received standard or FBR-enriched diet from postnatal day 21 to 48-53, when behavioral tests and ex vivo neurochemical analyses were performed. Phosphorylation levels of DARPP-32 in response to social and nonsocial cues, as index of dopamine D1 receptor activation, levels of expression of PPARα, vesicular glutamatergic and GABAergic transporters, and postsynaptic density protein PSD-95 were analyzed by immunoblotting in selected brain regions. RESULTS FBR administration relieved social impairment and perseverative behavior in VPA-exposed male and female rats, but it was only effective on female stereotypies. Dopamine D1 receptor signaling triggered by social interaction in the nucleus accumbens shell was blunted in VPA-exposed rats, and it was rescued by FBR treatment only in males. VPA-exposed rats of both sexes exhibited an increased ratio of striatal excitatory over inhibitory synaptic markers that was normalized by FBR treatment. LIMITATIONS This study did not directly address the extent of motivational deficit in VPA-exposed rats and whether FBR administration restored the likely decreased motivation to operate for social reward. Future studies using operant behavior protocols will address this relevant issue. CONCLUSIONS The results support the involvement of impaired motivational mechanisms in ASD-like social deficits and suggest the rationale for a possible pharmacological treatment. Moreover, the study highlights sex-related differences in the expression of ASD-like symptoms and their differential responses to FBR treatment.
Collapse
Affiliation(s)
- Simona Scheggi
- Department Molecular and Developmental Medicine, University of Siena, Via Aldo Moro, 2, Siena, Italy.
| | - Francesca Guzzi
- Department Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Giulia Braccagni
- Department Molecular and Developmental Medicine, University of Siena, Via Aldo Moro, 2, Siena, Italy
| | - Maria Graziella De Montis
- Department Molecular and Developmental Medicine, University of Siena, Via Aldo Moro, 2, Siena, Italy
| | - Marco Parenti
- Department Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Carla Gambarana
- Department Molecular and Developmental Medicine, University of Siena, Via Aldo Moro, 2, Siena, Italy
| |
Collapse
|
38
|
Marcondes LA, Nachtigall EG, Zanluchi A, de Carvalho Myskiw J, Izquierdo I, Furini CRG. Involvement of medial prefrontal cortex NMDA and AMPA/kainate glutamate receptors in social recognition memory consolidation. Neurobiol Learn Mem 2020; 168:107153. [DOI: 10.1016/j.nlm.2019.107153] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/12/2019] [Accepted: 12/23/2019] [Indexed: 11/28/2022]
|
39
|
Lawande NV, Ujjainwala AL, Christian CA. A Single Test to Study Social Behavior and Repetitive Self-grooming in Mice. Bio Protoc 2020; 10:e3499. [PMID: 32699808 DOI: 10.21769/bioprotoc.3499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
The ability to recognize and interact with members of the same species is essential for social communication. Investigating the neural substrates of social interest and recognition may offer insights into the behavioral differences present in disorders affecting social behavior. Assays used to study social interest in rodents include the 3-chamber test, a partition test, and a social interaction test. Here, we present a single protocol that can be used to quantify the level of social interest displayed by mice, the ability to distinguish between different individual mice (social recognition), and the level of repetitive self-grooming displayed. In the first part of the protocol, a social habituation/dishabituation test, the time spent by a test mouse sniffing a stimulus mouse is quantified over 9 trials. In the first 8 interactions, the same stimulus mouse is used repeatedly; on the ninth trial, a novel stimulus mouse is presented. Intact social recognition is indicated by a progressive decrease in the investigation time over trials 1-8, and an increase in trial 9. The interval between each social trial is used to quantify self-grooming, a stereotyped repetitive behavior in mice. We also present a method for randomized, blinded analysis of these behaviors to increase rigor and reproducibility of results. Therefore, this single behavioral test enables ready assessment of phenotypes of both social and repetitive behaviors in an integrated manner in the same animals. This feature can be advantageous in understanding interactions between these behaviors and phenotypes in mouse models with genetic variants associated with autism and other neurodevelopmental or neuropsychiatric disorders, which are often characterized by these behavioral differences.
Collapse
Affiliation(s)
- Niraj V Lawande
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ammar L Ujjainwala
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Catherine A Christian
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
40
|
Arakawa H. Sensorimotor developmental factors influencing the performance of laboratory rodents on learning and memory. Behav Brain Res 2019; 375:112140. [PMID: 31401145 PMCID: PMC6741784 DOI: 10.1016/j.bbr.2019.112140] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 02/08/2023]
Abstract
Behavioral studies in animal models have advanced our knowledge of brain function and the neural mechanisms of human diseases. Commonly used laboratory rodents, such as mice and rats, provide a useful tool for studying the behaviors and mechanisms associated with learning and memory processes which are cooperatively regulated by multiple underlying factors, including sensory and motor performance and emotional/defense innate components. Each of these factors shows unique ontogeny and governs the sustainment of behavioral performance in learning tasks, and thus, understanding the integrative processes of behavioral development are crucial in the accurate interpretation of the functional meaning of learning and memory behaviors expressed in commonly employed behavioral test paradigms. In this review, we will summarize the major findings in the developmental processes of rodent behavior on the basis of the emergence of fundamental components for sustaining learning and memory behaviors. Briefly, most sensory modalities (except for vision) and motor abilities are functional at the juvenile stage, in which several defensive components, including active and passive defensive strategies and risk assessment behavior, emerge. Sex differences are detectable from the juvenile stage through adulthood and are considerable factors that influence behavioral tests. The test paradigms addressed in this review include associative learning (with an emphasis on fear conditioning), spatial learning, and recognition. This basic background information will aid in accurately performing behavioral studies in laboratory rodents and will therefore contribute to reducing inappropriate interpretations of behavioral data and further advance research on learning and memory in rodent models.
Collapse
Affiliation(s)
- Hiroyuki Arakawa
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn St. HSF2/S251, Baltimore, MD, 21201, USA.
| |
Collapse
|
41
|
Disrupting the endocannabinoid system in early adolescence negatively impacts sociability. Pharmacol Biochem Behav 2019; 188:172832. [PMID: 31778723 DOI: 10.1016/j.pbb.2019.172832] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/22/2019] [Accepted: 11/22/2019] [Indexed: 01/23/2023]
Abstract
Animal models suggest that the endocannabinoid system (eCS) helps regulate various aspects of social behavior, including play behavior and social reward, during adolescence. Properly tuned endocannabinoid signaling may be a critical developmental component in the emergence of normal adult sociability. In the current experiment, we attempted to pharmacologically disrupt endocannabinoid tone during early adolescence, and then measure the behavioral effects at two subsequent time points. 36 male and 36 female Long Evans rats received daily injections of one of three treatments between post-natal day (PND) 25-39: 1) vehicle treatment, 2) 0.4 mg/kg CP55,940 (a potent CB1/CB2 receptor agonist), or 3) 0.5 mg/kg AM251 (a CB1 receptor antagonist/inverse agonist). Both soon after treatment (PND 40-44) and several weeks later (PND 66-70), subjects were tested in an elevated plus maze (EPM) for anxiety and in a three-chambered apparatus for sociability. For the latter test, the number of entries into each chamber and the amount of time spent investigating each target were measured. Analyses revealed significant main effects of both sex and age on sociability: males expressed greater sociability compared to females, and sociability was higher in adolescence than adulthood. Most importantly, drug treatment (both CP55,940 and AM251) attenuated sociability in adolescence without having a significant effect on anxiety in the EPM. However, this effect did not persist into adulthood. These results indicate that pharmacological disruption of endocannabinoid tone - through either chronic agonism or antagonism of cannabinoid receptors - during early adolescence has a detrimental effect on sociability. This effect may be caused by transient, compensatory alterations in the eCS.
Collapse
|
42
|
Tsai HW, Franklin M, Armoskus C, Taniguchi S, Moder C, Trang K, Santacruz M, Milla A. Androgenic regulation of sexually dimorphic expression of RNA binding motif protein 48 in the developing mouse cortex and hippocampus. Int J Dev Neurosci 2019; 78:33-44. [PMID: 31400491 PMCID: PMC6897302 DOI: 10.1016/j.ijdevneu.2019.07.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/25/2019] [Accepted: 07/31/2019] [Indexed: 11/28/2022] Open
Abstract
To further reveal the molecular mechanism underlying sexual differentiation of the mouse cerebral cortex and hippocampus, we reanalyzed our previous microarray study with Gene Ontology (GO) term enrichment and found that the GO term "RNA binding" was over-represented among the 89 sexually dimorphic candidate genes. Thus, we selected 16 autosomal genes annotated to the term RNA binding and profiled their mRNA expression in the developing male and female mouse cortex/hippocampus. During the first three weeks after birth, sex differences in mRNA levels of Khdrbs2, Nanos2, Rbm48, and Tdrd3 were observed in the mouse cortex/hippocampus. Of these genes, only the female-biased expression of Rbm48 in neonates was abolished by prenatal exposure to testosterone propionate (TP), while postnatal treatment of TP three weeks after birth increased Rbm48 and Tdrd3 mRNA levels in both sexes. Regardless of sex, the postnatal cortex/hippocampus also showed a marked increase in the content of androgen receptor (Ar) and estrogen receptor β (Esr2), but a decrease in estrogen receptor α (Esr1) and aromatase (Cyp19a1), which might confer the different responses of Rbm48 to prenatal and postnatal TP. Our results suggest that androgen-regulated, sexually dimorphic Rbm48 expression might present a novel molecular mechanism by which perinatal androgens control development of sexual dimorphism in cortical and hippocampal structure and function.
Collapse
Affiliation(s)
- Houng-Wei Tsai
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Michael Franklin
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Chris Armoskus
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Saori Taniguchi
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Courtney Moder
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Kathy Trang
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Marilisa Santacruz
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| | - Allyson Milla
- Department of Biological Sciences, California State University Long Beach, Long Beach, CA 90840, USA
| |
Collapse
|
43
|
Nakazawa H, Suzuki Y, Ishikawa Y, Bando Y, Yoshida S, Shiosaka S. Impaired social discrimination behavior despite normal social approach by kallikrein-related peptidase 8 knockout mouse. Neurobiol Learn Mem 2019; 162:47-58. [PMID: 31103466 DOI: 10.1016/j.nlm.2019.04.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 04/02/2019] [Accepted: 04/28/2019] [Indexed: 12/28/2022]
Abstract
For social mammals, recognition of conspecifics and discrimination of each other (social memory) is crucial to living in a stable colony. Here, we investigated whether kallikrein-related peptidase 8 (KLK8)-neuregulin 1 (NRG1)-ErbB signaling is crucial for social discrimination behavior using the social discrimination three chamber behavioral test. Klk8 knockout mice (NRG1-deactivated mice) exhibited normal social approach but impaired social discrimination. Intraventricular injection of recombinant NRG1177-246 into Klk8 knockout mice reversed this impaired social discrimination. This study reveals that KLK8 is a key regulator of NRG1-ErbB signaling, which contributes to social discrimination behavior.
Collapse
Affiliation(s)
- Hitomi Nakazawa
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa, Hokkaido 078-8510, Japan.
| | - Yuka Suzuki
- Department of Systems Life Engineering, Maebashi Institute of Technology, Maebashi, Gunma 371-0816, Japan
| | - Yasuyuki Ishikawa
- Department of Systems Life Engineering, Maebashi Institute of Technology, Maebashi, Gunma 371-0816, Japan
| | - Yoshio Bando
- Department of Anatomy, Akita University Graduate School of Medicine, Akita, Akita 010-8543, Japan
| | - Shigetaka Yoshida
- Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Asahikawa, Hokkaido 078-8510, Japan
| | - Sadao Shiosaka
- Graduate School of Biological Science, Nara Institute of Science and Technology (NAIST), Ikoma, Nara 630-0192, Japan.
| |
Collapse
|
44
|
Liu T, Ma Y, Zhang R, Zhong H, Wang L, Zhao J, Yang L, Fan X. Resveratrol ameliorates estrogen deficiency-induced depression- and anxiety-like behaviors and hippocampal inflammation in mice. Psychopharmacology (Berl) 2019; 236:1385-1399. [PMID: 30607478 DOI: 10.1007/s00213-018-5148-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/11/2018] [Indexed: 12/22/2022]
Abstract
RATIONALE Resveratrol (RSV) has been indicated to exhibit beneficial effects on depression and anxiety treatment by suppression of inflammatory processes. Depression triggered by deficiency of estrogen and anxiety-like behaviors are associated with inflammation. The role of RSV in ovariectomized mice is unclear. OBJECTIVES We examine whether the RSV, a Sirt1 activator, alleviates ovariectomy-induced anxiety- and depression-like behaviors through the inhibition of inflammatory processes. METHODS Female C57BL/6J mice (6-8 weeks of age, 17-20 g) were ovariectomized and treated with RSV at a dose of 20 mg/kg for 2 weeks. Depression- and anxiety-like behaviors were compared with vehicle-injected control animals. Immunohistochemistry and qPCR were used to detect inflammation in the hippocampal region. RESULTS Ovariectomized mice were observed to suffer from anxiety- and depression-like behaviors. These effects were attenuated by treatment with RSV. Immunohistochemical staining results showed that RSV could reverse the increase of microglial activation in the hippocampal dentate gyrus. At a molecular level, RSV inhibited the activation of NLRP3 and NF-κB in the hippocampal region caused by deficiency of estrogen. CONCLUSIONS RSV suppressed the production of inflammatory cytokines by enhancing Sirt1 levels. Our findings indicated that RSV-induced Sirt1 activation counteracted estrogen deficiency-induced psychobehavioral changes via inhibition of inflammatory processes in the hippocampus. In anxiety and depression disorders, RSV is supposed to be an effective treatment for postmenopausal changes.
Collapse
Affiliation(s)
- Tianyao Liu
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
- Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Yuanyuan Ma
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Ruiyu Zhang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
- Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Hongyu Zhong
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Lian Wang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Jinghui Zhao
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Ling Yang
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Xiaotang Fan
- Department of Developmental Neuropsychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
45
|
Sex-Dependent Sensory Phenotypes and Related Transcriptomic Expression Profiles Are Differentially Affected by Angelman Syndrome. Mol Neurobiol 2019; 56:5998-6016. [PMID: 30706369 DOI: 10.1007/s12035-019-1503-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/21/2019] [Indexed: 10/27/2022]
Abstract
Angelman syndrome (AS) is a genetic disorder which entails autism, intellectual disability, lack of speech, motor deficits, and seizure susceptibility. It is caused by the lack of UBE3A protein expression, which is an E3-ubiquitin ligase. Despite AS equal prevalence in males and females, not much data on how sex affects the syndrome was reported. In the herein study, we thoroughly characterized many behavioral phenotypes of AS mice. The behavioral data acquired was analyzed with respect to sex. In addition, we generated a new mRNA sequencing dataset. We analyzed the coding transcriptome expression profiles with respect to the effects of genotype and sex observed in the behavioral phenotypes. We identified several neurobehavioral aspects, especially sensory perception, where AS mice either lack the male-to-female differences observed in wild-type littermates or even show opposed differences. However, motor phenotypes did not show male-to-female variation between wild-type (WT) and AS mice. In addition, by utilizing the mRNA sequencing, we identified genes and isoforms with expression profiles that mirror the sensory perception results. These genes are differentially regulated in the two sexes with inverse expression profiles in AS mice compared to WT littermates. Some of these are known pain-related and estrogen-dependent genes. The observed differences in sex-dependent neurobehavioral phenotypes and the differential transcriptome expression profiles in AS mice strengthen the evidence for molecular cross talk between Ube3a protein and sex hormone receptors or their elicited pathways. These interactions are essential for understanding Ube3a deletion effects, beyond its E3-ligase activity.
Collapse
|
46
|
Chiang MC, Huang AJ, Wintzer ME, Ohshima T, McHugh TJ. A role for CA3 in social recognition memory. Behav Brain Res 2018; 354:22-30. [DOI: 10.1016/j.bbr.2018.01.019] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/15/2018] [Accepted: 01/17/2018] [Indexed: 11/15/2022]
|
47
|
Perez EC, Rodgers SP, Inoue T, Pedersen SE, Leasure JL, Gaber MW. Olfactory Memory Impairment Differs by Sex in a Rodent Model of Pediatric Radiotherapy. Front Behav Neurosci 2018; 12:158. [PMID: 30116180 PMCID: PMC6084003 DOI: 10.3389/fnbeh.2018.00158] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 07/09/2018] [Indexed: 11/24/2022] Open
Abstract
Although an effective treatment for pediatric brain tumors, cranial radiation therapy (CRT) damages surrounding healthy tissue, thereby disrupting brain development. Animal models of pediatric CRT have primarily relied on visual tasks to assess cognitive impairment. Moreover, there has been a lack of sex comparisons as most research on the cognitive effects of pediatric CRT does not include females. Therefore, we utilized olfaction, an ethologically relevant sensory modality, to assess cognitive impairment in an animal model of CRT that included both male and female mice. Specifically, we used the novel odor recognition (NOdorR) task with social odors to test recognition memory, a cognitive parameter that has been associated with olfactory neurogenesis, a form of cellular plasticity damaged by CRT. In addition to odor recognition memory, olfactory ability or discrimination of non-social and social odors were assessed both acutely and 3 months after CRT. Magnetic resonance imaging (MRI) and histology were performed after behavioral testing to assess long-term damage by CRT. Long-term but not acute radiation-induced impairment in odor recognition memory was observed, consistent with delayed onset of cognitive impairment in human patients. Males showed greater exploration of social odors than females, but general exploration was not affected by irradiation. However, irradiated males had impaired odor recognition memory in adulthood, compared to non-irradiated males (or simply male controls). Female olfactory recognition memory, in contrast, was dependent on estrus stage. CRT damage was demonstrated by (1) histological evaluation of olfactory neurogenesis, which suggested a reduction in CRT versus control, and (2) imaging analyses which showed that the majority of brain regions were reduced in volume by CRT. Specifically, two regions involved in social odor processing (amygdala and piriform cortex) were damaged by cranial irradiation in males but not females, paralleling olfactory recognition findings.
Collapse
Affiliation(s)
- Emma C Perez
- Behavioral Neuroscience Lab, Department of Psychology, University of Houston, Houston, TX, United States.,Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States
| | - Shaefali P Rodgers
- Behavioral Neuroscience Lab, Department of Psychology, University of Houston, Houston, TX, United States
| | - Taeko Inoue
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States
| | - Steen E Pedersen
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States.,Department of Physiology and Biochemistry, Ross University School of Medicine, Roseau, Dominica
| | - J Leigh Leasure
- Behavioral Neuroscience Lab, Department of Psychology, University of Houston, Houston, TX, United States.,Department of Biology and Biochemistry, University of Houston, Houston, TX, United States
| | - M Waleed Gaber
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
48
|
Wickens MM, Bangasser DA, Briand LA. Sex Differences in Psychiatric Disease: A Focus on the Glutamate System. Front Mol Neurosci 2018; 11:197. [PMID: 29922129 PMCID: PMC5996114 DOI: 10.3389/fnmol.2018.00197] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/18/2018] [Indexed: 12/21/2022] Open
Abstract
Alterations in glutamate, the primary excitatory neurotransmitter in the brain, are implicated in several psychiatric diseases. Many of these psychiatric diseases display epidemiological sex differences, with either males or females exhibiting different symptoms or disease prevalence. However, little work has considered the interaction of disrupted glutamatergic transmission and sex on disease states. This review describes the clinical and preclinical evidence for these sex differences with a focus on two conditions that are more prevalent in women: Alzheimer's disease and major depressive disorder, and three conditions that are more prevalent in men: schizophrenia, autism spectrum disorder, and attention deficit hyperactivity disorder. These studies reveal sex differences at multiple levels in the glutamate system including metabolic markers, receptor levels, genetic interactions, and therapeutic responses to glutamatergic drugs. Our survey of the current literature revealed a considerable need for more evaluations of sex differences in future studies examining the role of the glutamate system in psychiatric disease. Gaining a more thorough understanding of how sex differences in the glutamate system contribute to psychiatric disease could provide novel avenues for the development of sex-specific pharmacotherapies.
Collapse
Affiliation(s)
- Megan M Wickens
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, United States
| | - Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, United States.,Neuroscience Program, Temple University, Philadelphia, PA, United States
| | - Lisa A Briand
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, United States.,Neuroscience Program, Temple University, Philadelphia, PA, United States
| |
Collapse
|
49
|
Lian B, Gao J, Sui N, Feng T, Li M. Object, spatial and social recognition testing in a single test paradigm. Neurobiol Learn Mem 2018; 152:39-49. [PMID: 29778762 DOI: 10.1016/j.nlm.2018.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/29/2018] [Accepted: 05/16/2018] [Indexed: 12/27/2022]
Abstract
Animals have the ability to process information about an object or a conspecific's physical features and location, and alter its behavior when such information is updated. In the laboratory, the object, spatial and social recognition are often studied in separate tasks, making them unsuitable to study the potential dissociations and interactions among various types of recognition memories. The present study introduced a single paradigm to detect the object and spatial recognition, and social recognition of a familiar and novel conspecific. Specifically, male and female Sprague-Dawley adult (>75 days old) or preadolescent (25-28 days old) rats were tested with two objects and one social partner in an open-field arena for four 10-min sessions with a 20-min inter-session interval. After the first sample session, a new object replaced one of the sampled objects in the second session, and the location of one of the old objects was changed in the third session. Finally, a new social partner was introduced in the fourth session and replaced the familiar one. Exploration time with each stimulus was recorded and measures for the three recognitions were calculated based on the discrimination ratio. Overall results show that adult and preadolescent male and female rats spent more time exploring the social partner than the objects, showing a clear preference for social stimulus over nonsocial one. They also did not differ in their abilities to discriminate a new object, a new location and a new social partner from a familiar one, and to recognize a familiar conspecific. Acute administration of MK-801 (a NMDA receptor antagonist, 0.025 and 0.10 mg/kg, i.p.) after the sample session dose-dependently reduced the total time spent on exploring the social partner and objects in the adult rats, and had a significantly larger effect in the females than in the males. MK-801 also dose-dependently increased motor activity. However, it did not alter the object, spatial and social recognitions. These findings indicate that the new triple recognition paradigm is capable of recording the object, spatial location and social recognition together and revealing potential sex and age differences. This paradigm is also useful for the study of object and social exploration concurrently and can be used to evaluate cognition-altering drugs in various stages of recognition memories.
Collapse
Affiliation(s)
- Bin Lian
- Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, China; Faculty of Psychology, Southwest University, Chongqing, China
| | - Jun Gao
- Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, China; Faculty of Psychology, Southwest University, Chongqing, China
| | - Nan Sui
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Tingyong Feng
- Key Laboratory of Cognition and Personality (Southwest University), Ministry of Education, China; Faculty of Psychology, Southwest University, Chongqing, China.
| | - Ming Li
- Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE 68588-0308, USA; Faculty of Psychology, Southwest University, Chongqing, China.
| |
Collapse
|
50
|
Xing R, Liu F, Yang Y, Cui X, Wang T, Xie L, Zhao Y, Fang L, Yi T, Zheng B, Liu M, Chen H. GPR54 deficiency reduces the Treg population and aggravates experimental autoimmune encephalomyelitis in mice. SCIENCE CHINA-LIFE SCIENCES 2018; 61:675-687. [PMID: 29931449 DOI: 10.1007/s11427-017-9269-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 10/17/2017] [Indexed: 12/20/2022]
Abstract
GPR54 is highly expressed in the central nervous system and plays a crucial role in pubertal development. However, GRP54 is also expressed in the immune system, implying possible immunoregulatory functions. Here we investigated the role of GPR54 in T cell and immune tolerance. GPR54 deficiency led to an enlarged thymus, an increased number of thymocytes, and altered thymic micro-architecture starting around puberty, indicating GPR54 function in T-cell development through its regulatory effect on the gonadal system. However, flow cytometry revealed a significant reduction in the peripheral regulatory T cell population and a moderate decrease in CD4 single-positive thymocytes in prepubertal Gpr54-/- mice. These phenotypes were confirmed in chimeric mice with GPR54 deficient bone marrow-derived cells. In addition, we found elevated T cell activation in peripheral and thymic T cells in Gpr54-/- mice. When intact mice were immunized with myelin oligodendrocyte glycoprotein, a more severe experimental autoimmune encephalomyelitis (EAE) developed in the Gpr54-/- mice. Interestingly, aggravated EAE disease was also manifested in castrated and bone marrow chimeric Gpr54-/- mice compared to the respective wild-type control, suggesting a defect in self-tolerance resulting from GPR54 deletion through a mechanism that bypassed sex hormones. These findings demonstrate a novel role for GPR54 in regulating self-tolerant immunity in a sex hormone independent manner.
Collapse
MESH Headings
- Animals
- Disease Susceptibility
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Gene Expression
- Immune Tolerance/immunology
- Lymphocyte Activation/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myelin-Oligodendrocyte Glycoprotein/administration & dosage
- Myelin-Oligodendrocyte Glycoprotein/toxicity
- Receptors, Kisspeptin-1/deficiency
- Receptors, Kisspeptin-1/genetics
- Receptors, Kisspeptin-1/physiology
- Spleen/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocytes, Regulatory/immunology
- Thymus Gland/immunology
Collapse
Affiliation(s)
- Roumei Xing
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Fang Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yiqing Yang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xueqin Cui
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Tongtong Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Ling Xie
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yongliang Zhao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Lei Fang
- Third Venture Biotechnology Co., Ltd., Nanjing, 210042, China
| | - Tingfang Yi
- Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas, 77030, USA
| | - Biao Zheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Houston, Texas, 77030, USA.
| | - Huaqing Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|