1
|
Hernández-Vivanco A, de la Vega-Ruiz R, Montes-Mellado A, Azcoitia Í, Méndez P. Activational and organizational effects of sex hormones on hippocampal inhibitory neurons. J Neurosci 2025; 45:e1764242025. [PMID: 40097180 PMCID: PMC12044036 DOI: 10.1523/jneurosci.1764-24.2025] [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: 09/17/2024] [Revised: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 03/19/2025] Open
Abstract
Peripheral and brain-produced sex hormones exert sex-specific regulation of hippocampal cognitive function. Estrogens produced by neuronal aromatase regulate inhibitory neurons (INs) and hippocampal-dependent memory in adult female mice, but not in males. How and when this sex effect is established and how peripheral and brain sources of estrogens interact in the control of hippocampal INs is currently unknown. Using ex-vivo electrophysiology, fiber photometry, molecular analysis, estrous cycle monitoring and neonatal hormonal manipulations, we unveil estrous cycle dependent and independent features of CA1 Parvalbumin (PV) INs and hippocampal inhibition in adult female mice. Before puberty, aromatase is expressed in PV INs and regulates synaptic inhibition in female but not in male mice. Neonatal testosterone administration altered prepubertal female mouse hippocampus-dependent memory, PV IN function and estrogenic regulation of adult female synaptic inhibition and PV IN perineuronal nets. Our results suggest that sex differences in brain-derived estrogen regulation of CA1 inhibition are established by organizational effects of neonatal gonadal hormones and highlight the role of INs as mediators of the sexual differentiation of the hippocampus.Significance statement The actions of sex hormones on the hippocampus, a brain region involved in memory, differ between males and females but how and when these differences are established is not known. Our work identifies a population of hippocampal inhibitory neurons (INs) that are sensitive to hormonal fluctuations associated with the female estrous cycle. INs may produce estrogen, the main female sex hormone, before the onset of adult gonadal production (puberty). Brain-produced estrogen regulates female, but not male, juvenile INs, an effect that is abolished by a neonatal surge of testosterone that typically occurs in males around birth. Thus, early in life, sex hormones impact IN function suggesting a role for this neuronal population in the sexual differentiation of the hippocampus.
Collapse
Affiliation(s)
| | | | | | - Íñigo Azcoitia
- Department of Cell Biology, Universidad Complutense de Madrid, Madrid 28040, Spain
| | | |
Collapse
|
2
|
De Introna M, Krashia P, Sabetta A, La Barbera L, Nobili A, D'Amelio M, Cecconi F, Ammassari-Teule M, Pignataro A. Chemogenetic induction of CA1 hyperexcitability triggers indistinguishable autistic traits in asymptomatic mice differing in Ambra1 expression and sex. Transl Psychiatry 2025; 15:82. [PMID: 40097399 PMCID: PMC11914586 DOI: 10.1038/s41398-025-03271-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 12/18/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025] Open
Abstract
Among the genomic alterations identified as risk factors in mice models of autism spectrum disorders (ASD), heterozygous deletion of Ambra1 (Activating Molecule in Beclin1-Regulated Autophagy) triggers an ASD phenotype associated with hippocampal hyperexcitability exclusively in the female sex although Ambra1 protein is comparably expressed in the hippocampus of symptomatic females and asymptomatic males. Given the intricate relationship between Ambra1 deficiency and sex in the etiology of ASD, we took advantage of asymptomatic mice including Ambra1+/- males and wild-type (Wt) mice of both sexes to investigate whether their non-pathogenic variations in Ambra1 levels could underlie a differential susceptibility to exhibit ASD-like traits in response to experimental elevation of hippocampal excitability. Here we report that selective activation of inhibitory DREADD in CA1 parvalbumin-positive interneurons (PV-IN) reduces GABAergic currents onto pyramidal neurons (PN), causes social and attentional deficits, and augments the proportion of immature/thin spines in CA1 PN dendrites to the same extent in Ambra1+/- males and Wt mice of both sexes. Our findings show that the substantial hippocampal variations in pro-autophagic Ambra1 gene product shown by asymptomatic mice differing in mutation and/or sex do not underlie a differential reactivity to chemogenetic induction of idiopathic ASD.
Collapse
Affiliation(s)
- Margherita De Introna
- IRCCS Santa Lucia Foundation, Centro Europeo di Ricerca sul Cervello CERC, Rome, Italy
- Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy
| | - Paraskevi Krashia
- IRCCS Santa Lucia Foundation, Centro Europeo di Ricerca sul Cervello CERC, Rome, Italy
- Università Campus Bio-Medico di Roma, Rome, Italy
| | - Annamaria Sabetta
- IRCCS Santa Lucia Foundation, Centro Europeo di Ricerca sul Cervello CERC, Rome, Italy
- Dipartimento di Medicina Traslazionale e di Precisione, Sapienza Università di Roma, Rome, Italy
| | - Livia La Barbera
- IRCCS Santa Lucia Foundation, Centro Europeo di Ricerca sul Cervello CERC, Rome, Italy
- Università Campus Bio-Medico di Roma, Rome, Italy
| | - Annalisa Nobili
- IRCCS Santa Lucia Foundation, Centro Europeo di Ricerca sul Cervello CERC, Rome, Italy
- Università Campus Bio-Medico di Roma, Rome, Italy
| | - Marcello D'Amelio
- IRCCS Santa Lucia Foundation, Centro Europeo di Ricerca sul Cervello CERC, Rome, Italy
- Università Campus Bio-Medico di Roma, Rome, Italy
| | - Francesco Cecconi
- Fondazione Policlinico Universitario A. Gemelli, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Annabella Pignataro
- IRCCS Santa Lucia Foundation, Centro Europeo di Ricerca sul Cervello CERC, Rome, Italy.
- Institute of Translational Pharmacology, National Research Council, CNR, Rome, Italy.
| |
Collapse
|
3
|
Gilfarb RA, Ranade S, Dybas E, Biddle A, Stewart M, Rajesh A, Leuner B, Lenz KM. Hormonal contraceptives in adolescence impact the neuroimmune environment of the medial prefrontal cortex and hippocampus in female rats. Brain Behav Immun 2025; 127:315-328. [PMID: 39978694 DOI: 10.1016/j.bbi.2025.02.016] [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: 10/05/2024] [Revised: 02/02/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
Adolescence is a period of protracted neurodevelopment, during which the prefrontal cortex (PFC) undergoes significant remodeling. Microglia are integral to neurodevelopment and are sensitive to gonadal hormones, which increase during adolescence. Microglia and gonadal hormones can interact to influence adolescent development of the PFC (or medial prefrontal cortex [mPFC] in rodents). In females, gonadal hormones can be perturbed by using hormonal contraceptives (HCs). We predicted that HC administration over adolescence could affect microglia, other immunocompetent cells, and the neuroimmune environment of the developing mPFC. We also assessed HC effects on neuroimmune measures in the hippocampus, as the hippocampus also matures throughout adolescence and is sensitive to ovarian hormones. Intact post-pubertal female Sprague-Dawley rats received daily subcutaneous injections of vehicle or 10 ug ethinyl estradiol + 20 ug levonorgestrel (HCs) throughout adolescence from postnatal day (PND) 35-56. On PND 57 or 58, brains were collected for immunohistochemistry and qPCR. In the mPFC, HC-treated rats showed less Iba1 (microglia) immunolabeling and fewer Iba1+ cells. HC treatment also altered microglia morphology and reduced the spacing between microglia in the mPFC. In the hippocampus, HC-treated rats had reduced Iba1 immunolabeling in the dorsal CA1 and reductions in microglial cell complexity in dorsal CA1, ventral CA1, and ventral CA3. There were no effects of HCs on GFAP (astrocyte) immunolabeling in the mPFC or on astrocytes in any hippocampal subregion analyzed, except an increase in astrocyte number in the dorsal dentate gyrus. mPFC expression of genes related to phagocytosis (Cd68, Trem2) and neuroimmune signaling (Cx3cr1, Cx3cl1) were reduced in rats treated with HCs, but no gene expression changes were seen in the hippocampus. These data provide the first evidence that HCs given during the critical developmental period of adolescence can affect microglia properties in limbic brain regions.
Collapse
Affiliation(s)
- Rachel A Gilfarb
- Neuroscience Graduate Program, 460 Medical Center Drive, The Ohio State University, Columbus, OH 43210, USA
| | - Sanjana Ranade
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Elizabeth Dybas
- Neuroscience Graduate Program, 460 Medical Center Drive, The Ohio State University, Columbus, OH 43210, USA
| | - Abigail Biddle
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Meredith Stewart
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Abhishek Rajesh
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA
| | - Benedetta Leuner
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, 460 W. 12th Ave, Columbus, OH 43210, The Ohio State University, USA
| | - Kathryn M Lenz
- Department of Psychology, 1835 Neil Avenue, The Ohio State University, Columbus, OH 43210, USA; Institute for Behavioral Medicine Research, 460 Medical Center Drive, The Ohio State University, Columbus, OH 43210, USA; Department of Neuroscience, 460 W. 12th Ave, Columbus, OH 43210, The Ohio State University, USA.
| |
Collapse
|
4
|
van den Buuse M, Sun J, Gogos A. The effect of ovariectomy, 17β-estradiol treatment, and progesterone treatment on dopaminergic regulation of prepulse inhibition in adult and adolescent female mice. Horm Behav 2025; 167:105673. [PMID: 39721459 DOI: 10.1016/j.yhbeh.2024.105673] [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: 06/06/2024] [Revised: 10/04/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024]
Abstract
The aim of the present study was to investigate the role of ovarian hormones on dopaminergic regulation of prepulse inhibition (PPI), a measure of sensorimotor gating deficient in schizophrenia and other psychiatric illnesses. Either in adulthood (11 weeks of age) or adolescence (5 weeks of age), female mice underwent ovariectomy (OVX) and were implanted with 17β-estradiol, progesterone, or a combination of these hormones. All mice were tested in adulthood for the acute effect of the dopamine receptor agonist, apomorphine, on PPI. Apomorphine treatment reduced PPI in intact mice and this effect was blocked after OVX in adulthood. A low dose implant of 17β-estradiol prevented this OVX effect and reinstated apomorphine-induced PPI disruption. Following adolescent OVX, the effect of apomorphine was not altered and it significantly reduced PPI in adulthood. A low dose implant of 17β-estradiol following adolescent OVX effect blocked apomorphine-induced PPI disruption in adulthood. Apomorphine had no effect on PPI in any of the mice treated with the high dose of 17β-estradiol or a combination of low-dose 17β-estradiol and progesterone, irrespective of treatment age, suggesting an antipsychotic action. Apomorphine tended to disrupt PPI in mice treated with progesterone only, irrespective of age of OVX. These results suggest that in adult mice, circulating 17β-estradiol and progesterone play an important role in dopaminergic regulation of PPI. This role may develop during adolescence as similar effects of OVX and ovarian hormones were not observed following interventions in 5-week old mice. Our results also confirm and extend previous evidence that 17β-estradiol may have antipsychotic properties.
Collapse
Affiliation(s)
| | - Jenny Sun
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Andrea Gogos
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| |
Collapse
|
5
|
Di Miceli M, Rossitto M, Martinat M, Marchaland F, Kharbouche S, Graland M, Younes F, Séré A, Aubert A, Khabbaz LR, Madore C, Delpech JC, Martín R, Layé S. Modified neuroimmune processes and emotional behaviour in weaned and late adolescent male and female mice born via caesarean section. Sci Rep 2024; 14:29807. [PMID: 39616177 PMCID: PMC11608364 DOI: 10.1038/s41598-024-80770-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 11/21/2024] [Indexed: 02/07/2025] Open
Abstract
Elective and emergency Caesarean section (C-section) procedures are on the rise, exceeding the recommended guidelines by the World Health Organization. Higher morbidities and long-term health conditions are correlated to C-section deliveries, including neurodevelopmental disorders. During C-section delivery, newborns are not exposed to the vaginal commensal flora, which impedes the early establishment of the gut microbiota. The latter is essential for adequate neuro-immune processes to take place during infancy. In this study, we used a validated model of mice born by C-section (CSD), which mimics clinical observations of dysregulated gut microbiota. Animals were either born naturally or by CSD, before being adopted by dams who underwent delivery within the 12 preceding hours. Behavioural analyses were conducted at post-natal day (PND) 21 and 55. Our results indicate that animals born by C-section present significantly higher body weight in late (PND40-P53) but not early adolescence (PND21-P27), compared to animals born by vaginal delivery (VD). Male animals delivered by C-section presented significantly lower exploration time of the novel arm in the Y Maze test at PND55. However, at PND21, abnormal social interaction was witnessed in male and female animals born by CSD, with significantly decreased time spent interacting during the social interaction test. At both PND21 and PND55, animals from both sexes born by C-section presented significantly decreased time spent in the open arm of the Elevated Plus Maze test, compared to control animals. We then measured the expression of genes associated to neuroimmune interactions (microglia phenotype), inflammatory mediators and lipids in several brain structures of VD and CSD mice at PND21 and PND55. At weaning, animals born by CSD presented altered microglia, inflammatory and lipid metabolism signatures, with increased expression of Cd36, Csf1r and Tnfα in different brain regions of males, but not in females. At PND64, Csf1r, Tmem119 as well as C3ar1 were significantly increased in males born by C-section, but not in females. In males born by vaginal delivery, the expression of Cd36 at PND64 was correlated to anxiety at PND55, whilst a correlation between the expression of Clec7a and the number of head dippings in the elevated plus maze was also noted in males born by CSD. Altogether, our study shows altered emotional behaviour in animals delivered by CSD, which is likely explained by underlying neuro-inflammatory processes in different brain regions. Our work further supports the long-term consequences of CSD on brain health.
Collapse
Affiliation(s)
- Mathieu Di Miceli
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France.
- Worcester Biomedical Research Group, School of Science and the Environment, University of Worcester, Henwick Grove, Worcester, WR2 6AJ, UK.
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| | - Moïra Rossitto
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Maud Martinat
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Flore Marchaland
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Sarah Kharbouche
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Marion Graland
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Farah Younes
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Alexandra Séré
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Agnès Aubert
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Lydia Rabbaa Khabbaz
- Laboratoire de Pharmacologie, Pharmacie Clinique et Contrôle de Qualité des Médicaments, Faculty of Pharmacy, Saint-Joseph University of Beirut, Beirut, Lebanon
| | - Charlotte Madore
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | | | - Rebeca Martín
- Micalis Institute, AgroParisTech, INRAE, Université Paris-Saclay, 78350, Jouy-En-Josas, France
| | - Sophie Layé
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| |
Collapse
|
6
|
Santos-Silva T, Souza BK, Colodete DAE, Campos LR, Lima TSA, Guimarães FS, Gomes FV. Differential Impact of Adolescent or Adult Stress on Behavior and Cortical Parvalbumin Interneurons and Perineuronal Nets in Male and Female Mice. Int J Neuropsychopharmacol 2024; 27:pyae042. [PMID: 39276147 PMCID: PMC11639180 DOI: 10.1093/ijnp/pyae042] [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: 05/24/2024] [Accepted: 09/12/2024] [Indexed: 09/16/2024] Open
Abstract
BACKGROUND Stress has become a common public health concern, contributing to the rising prevalence of psychiatric disorders. Understanding the impact of stress considering critical variables, such as age, sex, and individual differences, is of the utmost importance for developing effective intervention strategies. METHODS Stress effects (daily footshocks for 10 days) during adolescence (postnatal day [PND] 31-40) and adulthood (PND 65-74) were investigated on behavioral outcomes and parvalbumin (PV)-expressing GABAergic interneurons and their associated perineuronal nets (PNNs) in the prefrontal cortex of male and female mice 5 weeks post stress. RESULTS In adulthood, adolescent stress induced behavioral alterations in male mice, including anxiety-like behaviors, social deficits, cognitive impairments, and altered dopamine system responsivity. Applying integrated behavioral z-score analysis, we identified sex-specific differences in response to adolescent stress, with males displaying greater vulnerability than females. Furthermore, adolescent-stressed male mice showed decreased PV+ and PNN+ cell numbers and PV+/PNN+ colocalization, while in females, adolescent stress reduced prefrontal PV+/PNN+ colocalization in the prefrontal cortex. Further analysis identified distinct behavioral clusters, with certain females demonstrating resilience to adolescent stress-induced deficits in sociability and PV+ cell number. Adult stress in male and female mice did not cause long-lasting changes in behavior and PV+ and PNN+ cell number. CONCLUSION Our findings indicate that the timing of stress, sex, and individual variabilities seem to be determinants for the development of behavioral changes associated with psychiatric disorders, particularly in male mice during adolescence.
Collapse
Affiliation(s)
- Thamyris Santos-Silva
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Beatriz Kinchin Souza
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Débora Akemi Endo Colodete
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Lara Ramos Campos
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Thaís Santos Almeida Lima
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Francisco S Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Felipe V Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
7
|
Uliana DL, Lisboa JRF, Gomes FV, Grace AA. The excitatory-inhibitory balance as a target for the development of novel drugs to treat schizophrenia. Biochem Pharmacol 2024; 228:116298. [PMID: 38782077 PMCID: PMC11410545 DOI: 10.1016/j.bcp.2024.116298] [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: 02/01/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
The intricate balance between excitation and inhibition (E/I) in the brain plays a crucial role in normative information processing. Dysfunctions in the E/I balance have been implicated in various psychiatric disorders, including schizophrenia (SCZ). In particular, abnormalities in GABAergic signaling, specifically in parvalbumin (PV)-containing interneurons, have been consistently observed in SCZ pathophysiology. PV interneuron function is vital for maintaining an ideal E/I balance, and alterations in PV interneuron-mediated inhibition contribute to circuit deficits observed in SCZ, including hippocampus hyperactivity and midbrain dopamine system overdrive. While current antipsychotic medications primarily target D2 dopamine receptors and are effective primarily in treating positive symptoms, novel therapeutic strategies aiming to restore the E/I balance could potentially mitigate not only positive symptoms but also negative symptoms and cognitive deficits. This could involve, for instance, increasing the inhibitory drive onto excitatory neurons or decreasing the putative enhanced pyramidal neuron activity due to functional loss of PV interneurons. Compounds targeting the glycine site at glutamate NMDA receptors and muscarinic acetylcholine receptors on PV interneurons that can increase PV interneuron drive, as well as drugs that increase the postsynaptic action of GABA, such as positive allosteric modulators of α5-GABA-A receptors, and decrease glutamatergic output, such as mGluR2/3 agonists, represent promising approaches. Preventive strategies aiming at E/I balance also represent a path to reduce the risk of transitioning to SCZ in high-risk individuals. Therefore, compounds with novel mechanisms targeting E/I balance provide optimism for more effective and tailored interventions in the management of SCZ.
Collapse
Affiliation(s)
- Daniela L Uliana
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joao Roberto F Lisboa
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Felipe V Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
8
|
Fournier LA, Phadke RA, Salgado M, Brack A, Nocon JC, Bolshakova S, Grant JR, Padró Luna NM, Sen K, Cruz-Martín A. Overexpression of the schizophrenia risk gene C4 in PV cells drives sex-dependent behavioral deficits and circuit dysfunction. iScience 2024; 27:110800. [PMID: 39310747 PMCID: PMC11416532 DOI: 10.1016/j.isci.2024.110800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/09/2024] [Accepted: 08/20/2024] [Indexed: 09/25/2024] Open
Abstract
Fast-spiking parvalbumin (PV)-positive cells are key players in orchestrating pyramidal neuron activity, and their dysfunction is consistently observed in myriad brain diseases. To understand how immune complement pathway dysregulation in PV cells drives disease pathogenesis, we have developed a transgenic line that permits cell-type specific overexpression of the schizophrenia-associated C4 gene. We found that overexpression of mouse C4 (mC4) in PV cells causes sex-specific alterations in anxiety-like behavior and deficits in synaptic connectivity and excitability of PFC PV cells. Using a computational model, we demonstrated that these microcircuit deficits led to hyperactivity and disrupted neural communication. Finally, pan-neuronal overexpression of mC4 failed to evoke the same deficits in behavior as PV-specific mC4 overexpression, suggesting that perturbations of this neuroimmune gene in fast-spiking neurons are especially detrimental to circuits associated with anxiety-like behavior. Together, these results provide a causative link between C4 and the vulnerability of PV cells in brain disease.
Collapse
Affiliation(s)
- Luke A. Fournier
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Rhushikesh A. Phadke
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
| | - Maria Salgado
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
| | - Alison Brack
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
| | - Jian Carlo Nocon
- Neurophotonics Center, Boston University, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
- Hearing Research Center, Boston University, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sonia Bolshakova
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
- Bioinformatics MS Program, Boston University, Boston, MA, USA
| | - Jaylyn R. Grant
- Biological Sciences, Eastern Illinois University, Charleston, IL, USA
- The Summer Undergraduate Research Fellowship (SURF) Program, Boston University, Boston, MA, USA
| | - Nicole M. Padró Luna
- The Summer Undergraduate Research Fellowship (SURF) Program, Boston University, Boston, MA, USA
- Biology Department, College of Natural Sciences, University of Puerto Rico, Rio Piedras Campus, San Juan, PR, USA
| | - Kamal Sen
- Neurophotonics Center, Boston University, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
- Hearing Research Center, Boston University, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Alberto Cruz-Martín
- Neurobiology Section in the Department of Biology, Boston University, Boston, MA, USA
- Molecular Biology, Cell Biology & Biochemistry Program, Boston University, Boston, MA, USA
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- NeuroTechnology Center (NTC), University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
9
|
Méndez P, de la Vega-Ruiz R, Montes-Mellado A. Estrogenic regulation of hippocampal inhibitory system across lifespan. J Neuroendocrinol 2024:e13441. [PMID: 39143852 DOI: 10.1111/jne.13441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/08/2024] [Accepted: 08/01/2024] [Indexed: 08/16/2024]
Abstract
Estrogens produced in peripheral tissues and locally in the brain are potent neuromodulators. The function of the hippocampus, a brain region essential for episodic memory and spatial navigation, relies on the activity of ensembles of excitatory neurons whose activity is temporally and spatially coordinated by a wide diversity of inhibitory neurons (INs) types. Over the last years, we have accumulated evidence that indicates that estrogens regulate the function of hippocampal INs through different mechanisms, including transcriptional regulation and rapid nongenomic signaling. Here, we argue that the well-documented influence of estrogens on episodic memory may be related to the actions of local and peripheral estrogens on the heterogenous populations of hippocampal INs. We discuss how physiological changes in peripheral sex hormone levels throughout lifespan may interact with local brain sources to regulate IN function at different stages of life, from early hippocampal development to the aging brain. We conclude that considering INs as mediators of sex hormone actions in the hippocampus across the healthy life span will benefit our understanding of sex-biased neurodevelopmental disorders and physiological aging.
Collapse
|
10
|
Van Zandt M, Flanagan D, Pittenger C. Sex differences in the distribution and density of regulatory interneurons in the striatum. Front Cell Neurosci 2024; 18:1415015. [PMID: 39045533 PMCID: PMC11264243 DOI: 10.3389/fncel.2024.1415015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/25/2024] [Indexed: 07/25/2024] Open
Abstract
Introduction Dysfunction of the cortico-basal circuitry - including its primary input nucleus, the striatum - contributes to neuropsychiatric disorders, such as autism and Tourette Syndrome (TS). These conditions show marked sex differences, occurring more often in males than in females. Regulatory interneurons, such as cholinergic interneurons (CINs) and parvalbumin-expressing GABAergic fast spiking interneurons (FSIs), are implicated in human neuropsychiatric disorders such as TS, and ablation of these interneurons produces relevant behavioral pathology in male mice, but not in females. Here we investigate sex differences in the density and distribution of striatal interneurons. Methods We use stereological quantification of CINs, FSIs, and somatostatin-expressing (SOM) GABAergic interneurons in the dorsal striatum (caudate-putamen) and the ventral striatum (nucleus accumbens) in male and female mice. Results Males have a higher density of CINs than females, especially in the dorsal striatum; females have equal distribution between dorsal and ventral striatum. FSIs showed similar distributions, with a greater dorsal-ventral density gradient in males than in females. SOM interneurons were denser in the ventral than in the dorsal striatum, with no sex differences. Discussion These sex differences in the density and distribution of FSIs and CINs may contribute to sex differences in basal ganglia function, particularly in the context of psychopathology.
Collapse
Affiliation(s)
- Meghan Van Zandt
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Deirdre Flanagan
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
| | - Christopher Pittenger
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States
- Yale Child Study Center, Yale University School of Medicine, New Haven, CT, United States
- Department of Psychology, Yale School of Arts and Sciences, New Haven, CT, United States
- Center for Brain and Mind Health, Yale University School of Medicine, New Haven, CT, United States
- Wu-Tsai Institute, Yale University, New Haven, CT, United States
| |
Collapse
|
11
|
Jana S, Giri B, Das S, Manna A, Mandal SC, Ranjan Jana N. Azadiradione up-regulates the expression of parvalbumin and BDNF via Ube3a. Gene 2024; 897:148081. [PMID: 38101713 DOI: 10.1016/j.gene.2023.148081] [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: 11/28/2023] [Accepted: 12/12/2023] [Indexed: 12/17/2023]
Abstract
Azadiradione is a small bioactive limonoid found in the seed of Azadirachta Indica, an Indian medicinal plant commonly known as Neem. Recently, it has been shown to ameliorate the disease pathology in fly and mouse model of Huntington's disease by restoring impaired proteostasis. Here we report that the azadiradione could be involved in modulating the synaptic function through increased expression of Ube3a, a dual function protein having ubiquitin ligase and co-activator functions and associated with Angelman syndrome and autism. Treatment of azadiradione to HT22 hippocampal cell line and in adult mice induced the expression of Ube3a as well as two important synaptic function and plasticity regulating proteins, parvalbumin and brain-derived neurotropic factor (BDNF). Interestingly, another synaptic plasticity modulating protein Arc (activity-regulated cytoskeletal associated protein) was down-regulated by azadiradione. Partial knockdown of Ube3a in HT22 cell abrogated azadiradione induced expression of parvalbumin and BDNF. Ube3a-maternal deficient mice also exhibited significantly decreased expression of parvalbumin and BDNF in their brain and treatment of azadiradione in these animals did not rescue the altered expression of either parvalbumin or BDNF. These results indicate that azadiradione-induced expression of parvalbumin and BDNF in the brain is mediated through Ube3a and suggest that azadiradione could be implicated in restoring synaptic dysfunction in many neuropsychiatric/neurodegenerative disorders.
Collapse
Affiliation(s)
- Sudipta Jana
- School of Bioscience, Indian Institute of Technology, Kharagpur 721302, India
| | - Bhaskarjyoti Giri
- School of Bioscience, Indian Institute of Technology, Kharagpur 721302, India
| | - Sagarika Das
- School of Bioscience, Indian Institute of Technology, Kharagpur 721302, India
| | - Anirban Manna
- Division of Molecular Medicine, Bose Institute, Kolkata 700054, India
| | - Subhash C Mandal
- Division of Pharmacognosy, Department of Pharmaceutical Technology, Jadavpur University, Jadavpur, Kolkata 700032, India
| | - Nihar Ranjan Jana
- School of Bioscience, Indian Institute of Technology, Kharagpur 721302, India.
| |
Collapse
|
12
|
Ortelli OA, Pitcairn SR, Dyson CH, Weiner JL. Sexually dimorphic effects of a modified adolescent social isolation paradigm on behavioral risk factors of alcohol use disorder in Long Evans Rats. ADDICTION NEUROSCIENCE 2023; 9:100134. [PMID: 38188062 PMCID: PMC10768969 DOI: 10.1016/j.addicn.2023.100134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Early life stress (ELS) is a major risk factor for alcohol use disorder (AUD) and comorbid neuropsychiatric conditions. We previously demonstrated that an adolescent social isolation (aSI) model of ELS significantly increased behavioral risk factors for these disorders (e.g. anxiety-like behaviors, alcohol drinking) in male, but not female rats. Since many neurodevelopmental milestones are accelerated in females, we investigated whether an earlier/shorter isolation window (PND 21-38) would yield comparable phenotypes in both sexes. In two experiments, Long Evans rats were socially isolated (SI) or group-housed (GH) on postnatal day (PND) 21 and locomotion was assessed in the open field test (OFT; PND 30). Experiment 1 also assessed behavior on the elevated plus-maze (EPM) (PND 32). In Experiment 2, all rats were single housed on PND 38 to assess home cage alcohol drinking. Experiment 1 revealed that SI females had increased locomotor activity in the OFT but did not differ from GH subjects on the EPM. The OFT results were replicated in both sexes in Experiment 2 and both male and female SI rats had significantly greater ethanol consumption during an eight day continuous access paradigm. In contrast, during subsequent intermittent two-bottle choice drinking, only SI females displayed greater ethanol intake and preference and increased consumption of a quinine-adulterated alcohol solution. These findings demonstrate that early life social isolation can promote AUD vulnerability-related phenotypes in female rats but that there are profound sex differences in the vulnerability window to this early life stressor. Uncovering the neural mechanisms responsible for these sexually dimorphic differences in sensitivity to ELS may shed light on the biological substrates associated with vulnerability to AUD and comorbid disorders of negative emotion in men and women.
Collapse
Affiliation(s)
- Olivia A. Ortelli
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Stacy R. Pitcairn
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Christina H. Dyson
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Jeffrey L. Weiner
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
13
|
Pentz AB, Timpe CMF, Normann EM, Slapø NB, Melle I, Lagerberg TV, Steen NE, Westlye LT, Jönsson EG, Haukvik UK, Moberget T, Andreassen OA, Elvsåshagen T. Mismatch negativity in schizophrenia spectrum and bipolar disorders: Group and sex differences and associations with symptom severity. Schizophr Res 2023; 261:80-93. [PMID: 37716205 DOI: 10.1016/j.schres.2023.09.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/15/2023] [Accepted: 09/04/2023] [Indexed: 09/18/2023]
Abstract
OBJECTIVE Research increasingly implicates glutamatergic dysfunction in the pathophysiologies of psychotic disorders. Auditory mismatch negativity (MMN) is an electroencephalography (EEG) waveform linked to glutamatergic neurotransmission and is consistently attenuated in schizophrenia (SCZ). MMN consists of two subcomponents, the repetition positivity (RP) and deviant negativity (DN) possibly reflecting different neural mechanisms. However, whether MMN reduction is present across different psychotic disorders, linked to distinct symptom clusters, or related to sex remain to be clarified. METHODS Four hundred participants including healthy controls (HCs; n = 296) and individuals with SCZ (n = 39), bipolar disorder (BD) BD typeI (n = 35), or BD type II (n = 30) underwent a roving MMN paradigm and clinical evaluation. MMN, RP and DN as well their memory traces were recorded at the FCZ electrode. Analyses of variance and linear regression models were used both transdiagnostically and within clinical groups. RESULTS MMN was reduced in SCZ compared to BD (p = 0.006, d = 0.55) and to HCs (p < 0.001, d = 0.63). There was a significant group × sex interaction (p < 0.003) and the MMN impairment was only detected in males with SCZ. MMN amplitude correlated positively with Positive and Negative Syndrome Scale total score and negatively with Global Assessment of Functioning Scale score. The deviant negativity was impaired in males with SCZ. No group differences in memory trace indices of the MMN, DN, or RP. CONCLUSION MMN was attenuated in SCZ and correlated with greater severity of psychotic symptoms and lower level of functioning. Our results may indicate sex-dependent differences of glutamatergic function in SCZ.
Collapse
Affiliation(s)
- Atle Bråthen Pentz
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway.
| | - Clara Maria Fides Timpe
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | | | - Nora Berz Slapø
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway
| | - Ingrid Melle
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway
| | - Trine Vik Lagerberg
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway
| | - Nils Eiel Steen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway; Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Lars T Westlye
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | - Erik G Jönsson
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway; Centre for Psychiatric Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Sciences, Stockholm Region, Stockholm, Sweden
| | - Unn K Haukvik
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway; Department of Adult Psychiatry, Institute of Clinical Medicine, University of Oslo, Norway; Department of Forensic Psychiatry Research, Oslo University Hospital, Norway
| | - Torgeir Moberget
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway; Department of Psychology, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway
| | - Torbjørn Elvsåshagen
- NORMENT, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Norway; Department of Neurology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
14
|
Terstege DJ, Epp JR. Parvalbumin as a sex-specific target in Alzheimer's disease research - A mini-review. Neurosci Biobehav Rev 2023; 153:105370. [PMID: 37619647 DOI: 10.1016/j.neubiorev.2023.105370] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, and both the incidence of this disease and its associated cognitive decline disproportionally effect women. While the etiology of AD is unknown, recent work has demonstrated that the balance of excitatory and inhibitory activity across the brain may serve as a strong predictor of cognitive impairments in AD. Across the cortex, the most prominent source of inhibitory signalling is from a class of parvalbumin-expressing interneurons (PV+). In this mini-review, the impacts of sex- and age-related factors on the function of PV+ neurons are examined within the context of vulnerability to AD pathology. These primary factors of influence include changes in brain metabolism, circulating sex hormone levels, and inflammatory response. In addition to positing the increased vulnerability of PV+ neurons to dysfunction in AD, this mini-review highlights the critical importance of presenting sex stratified data in the study of AD.
Collapse
Affiliation(s)
- Dylan J Terstege
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta T2N 4N1, Canada
| | - Jonathan R Epp
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta T2N 4N1, Canada.
| |
Collapse
|
15
|
Woods R, Lorusso J, Fletcher J, ElTaher H, McEwan F, Harris I, Kowash H, D'Souza SW, Harte M, Hager R, Glazier JD. Maternal immune activation and role of placenta in the prenatal programming of neurodevelopmental disorders. Neuronal Signal 2023; 7:NS20220064. [PMID: 37332846 PMCID: PMC10273029 DOI: 10.1042/ns20220064] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
Maternal infection during pregnancy, leading to maternal immune activation (mIA) and cytokine release, increases the offspring risk of developing a variety of neurodevelopmental disorders (NDDs), including schizophrenia. Animal models have provided evidence to support these mechanistic links, with placental inflammatory responses and dysregulation of placental function implicated. This leads to changes in fetal brain cytokine balance and altered epigenetic regulation of key neurodevelopmental pathways. The prenatal timing of such mIA-evoked changes, and the accompanying fetal developmental responses to an altered in utero environment, will determine the scope of the impacts on neurodevelopmental processes. Such dysregulation can impart enduring neuropathological changes, which manifest subsequently in the postnatal period as altered neurodevelopmental behaviours in the offspring. Hence, elucidation of the functional changes that occur at the molecular level in the placenta is vital in improving our understanding of the mechanisms that underlie the pathogenesis of NDDs. This has notable relevance to the recent COVID-19 pandemic, where inflammatory responses in the placenta to SARS-CoV-2 infection during pregnancy and NDDs in early childhood have been reported. This review presents an integrated overview of these collective topics and describes the possible contribution of prenatal programming through placental effects as an underlying mechanism that links to NDD risk, underpinned by altered epigenetic regulation of neurodevelopmental pathways.
Collapse
Affiliation(s)
- Rebecca M. Woods
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jarred M. Lorusso
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jennifer Fletcher
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Heidi ElTaher
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
- Department of Physiology, Faculty of Medicine, Alexandria University, Egypt
| | - Francesca McEwan
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Isabella Harris
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Hager M. Kowash
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9WL, U.K
| | - Stephen W. D'Souza
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9WL, U.K
| | - Michael Harte
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Medicine, Biology and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Reinmar Hager
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| | - Jocelyn D. Glazier
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, U.K
| |
Collapse
|
16
|
Woodward EM, Ringland A, Ackerman J, Coutellier L. Prepubertal ovariectomy confers resilience to stress-induced anxiety in adult female mice. Psychoneuroendocrinology 2023; 148:105997. [PMID: 36470154 PMCID: PMC9898172 DOI: 10.1016/j.psyneuen.2022.105997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 11/29/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022]
Abstract
The increased vulnerability to stress-induced neuropsychiatric disorders in women, including anxiety disorders, does not emerge until pubertal onset, suggesting a role for ovarian hormones in organizing sex-specific vulnerability to anxiety. Parvalbumin (PV) interneurons in the prefrontal cortex are a potential target for these ovarian hormones. PV+ interneurons undergo maturation during the adolescent period and have been shown to be sensitive to stress and to mediate stress-induced anxiety in female mice. To test the idea that ovarian hormones at puberty are necessary for the acquisition of sensitivity to stress, hypothetically driving the response of PV+ interneurons to stress, we performed ovariectomy or sham surgery before pubertal onset in female mice. These mice then were exposed to four weeks of unpredictable chronic mild stress in adulthood. We then assessed anxiety-like behavior and PV/FosB colocalization in the medial PFC. Additionally, we assessed stress-induced anxiety-like behavior in female mice following ovariectomy in adulthood to determine if puberty is a sensitive period for ovarian hormones in mediating vulnerability to stress. We found that prepubertal ovariectomy protects against the development of anxiety-like behavior in adulthood, an effect not found following ovariectomy in adulthood. This effect may be independent of ovarian hormones on prefrontal PV+ interneurons response to stress.
Collapse
Affiliation(s)
- Emma M Woodward
- Department of Neuroscience, Ohio State University, 255 Institute for Behavioral Medicine Research Building, 460 Medical Center Drive, Columbus, OH 43210, USA.
| | - Amanda Ringland
- Department of Neuroscience, Ohio State University, 255 Institute for Behavioral Medicine Research Building, 460 Medical Center Drive, Columbus, OH 43210, USA; Department of Psychology, Ohio State University, 1835 Neil Avenue, Columbus, OH 43210, USA.
| | - Jennifer Ackerman
- Department of Neuroscience, Ohio State University, 255 Institute for Behavioral Medicine Research Building, 460 Medical Center Drive, Columbus, OH 43210, USA; Department of Psychology, Ohio State University, 1835 Neil Avenue, Columbus, OH 43210, USA.
| | - Laurence Coutellier
- Department of Neuroscience, Ohio State University, 255 Institute for Behavioral Medicine Research Building, 460 Medical Center Drive, Columbus, OH 43210, USA; Department of Psychology, Ohio State University, 1835 Neil Avenue, Columbus, OH 43210, USA.
| |
Collapse
|
17
|
Zhu X, Grace AA. Sex- and exposure age-dependent effects of adolescent stress on ventral tegmental area dopamine system and its afferent regulators. Mol Psychiatry 2023; 28:611-624. [PMID: 36224257 PMCID: PMC9918682 DOI: 10.1038/s41380-022-01820-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 11/09/2022]
Abstract
Adolescent stress is a risk factor for schizophrenia. Emerging evidence suggests that age-dependent sensitive windows for childhood trauma are associated more strongly with adult psychosis, but the neurobiological basis and potential sex differences are unknown.Using in vivo electrophysiology and immunohistology in rats, we systematically compared the effects of two age-defined adolescent stress paradigms, prepubertal (postnatal day [PD] 21-30; PreP-S) and postpubertal (PD41-50; PostP-S) foot-shock and restraint combined stress, on ventral tegmental area (VTA) dopaminergic activity, pyramidal neuron activity in the ventral hippocampus (vHipp) and the basolateral amygdala (BLA), corticoamygdalar functional inhibitory control, and vHipp and BLA parvalbumin interneuron (PVI) impairments. These endpoints were selected based on their well-documented roles in the pathophysiology of psychosis.Overall, we found distinct sex- and exposure age-dependent stress vulnerability. Specifically, while males were selectively vulnerable to PreP-S-induced adult VTA dopamine neuron and vHipp hyperactivities, females were selectively vulnerable to PostP-S. These male selective PreP-S effects were correlated with stress-induced aberrant persistent BLA hyperactivity, dysfunctional prefrontal inhibitory control of BLA neurons, and vHipp/BLA PVI impairments. In contrast, female PostP-S only produced vHipp PVI impairments in adults, with the BLA structure and functions largely unaffected.Our results indicated distinct adolescent-sensitive periods during which stress can sex-dependently confer maximal risks to corticolimbic systems to drive dopamine hyperactivity, which provide critical insights into the neurobiological basis for sex-biased stress-related psychopathologies emphasizing but not limited to schizophrenia. Furthermore, our work also provides a framework for future translational research on age-sensitive targeted interventions.
Collapse
Affiliation(s)
- Xiyu Zhu
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Anthony A Grace
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
18
|
Altered visual cortex excitability in premenstrual dysphoric disorder: Evidence from magnetoencephalographic gamma oscillations and perceptual suppression. PLoS One 2022; 17:e0279868. [PMID: 36584199 PMCID: PMC9803314 DOI: 10.1371/journal.pone.0279868] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 12/15/2022] [Indexed: 12/31/2022] Open
Abstract
Premenstrual dysphoric disorder (PMDD) is a psychiatric condition characterized by extreme mood shifts during the luteal phase of the menstrual cycle (MC) due to abnormal sensitivity to neurosteroids and unbalanced neural excitation/inhibition (E/I) ratio. We hypothesized that in women with PMDD in the luteal phase, these factors would alter the frequency of magnetoencephalographic visual gamma oscillations, affect modulation of their power by excitatory drive, and decrease perceptual spatial suppression. Women with PMDD and control women were examined twice-during the follicular and luteal phases of their MC. We recorded visual gamma response (GR) while modulating the excitatory drive by increasing the drift rate of the high-contrast grating (static, 'slow', 'medium', and 'fast'). Contrary to our expectations, GR frequency was not affected in women with PMDD in either phase of the MC. GR power suppression, which is normally associated with a switch from the 'optimal' for GR slow drift rate to the medium drift rate, was reduced in women with PMDD and was the only GR parameter that distinguished them from control participants specifically in the luteal phase and predicted severity of their premenstrual symptoms. Over and above the atypical luteal GR suppression, in both phases of the MC women with PMDD had abnormally strong GR facilitation caused by a switch from the 'suboptimal' static to the 'optimal' slow drift rate. Perceptual spatial suppression did not differ between the groups but decreased from the follicular to the luteal phase only in PMDD women. The atypical modulation of GR power suggests that neuronal excitability in the visual cortex is constitutively elevated in PMDD and that this E/I imbalance is further exacerbated during the luteal phase. However, the unaltered GR frequency does not support the hypothesis of inhibitory neuron dysfunction in PMDD.
Collapse
|
19
|
Banerjee T, Pati S, Tiwari P, Vaidya VA. Chronic hM3Dq-DREADD-mediated chemogenetic activation of parvalbumin-positive inhibitory interneurons in postnatal life alters anxiety and despair-like behavior in adulthood in a task- and sex-dependent manner. J Biosci 2022. [DOI: 10.1007/s12038-022-00308-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
20
|
Chinn GA, Duong K, Horovitz TR, Russell JMS, Sall JW. Testosterone is Sufficient to Impart Susceptibility to Isoflurane Neurotoxicity in Female Neonatal Rats. J Neurosurg Anesthesiol 2022; 34:429-436. [PMID: 34127616 PMCID: PMC8671561 DOI: 10.1097/ana.0000000000000786] [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: 03/08/2021] [Accepted: 05/10/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Volatile anesthetic exposure during development leads to long-term cognitive deficits in rats which are dependent on age and sex. Female rats are protected relative to male rats for the same exposure on postnatal day 7. Here we test our hypothesis that androgens can modulate chloride cotransporter expression to alter the susceptibility to neurotoxicity from GABAergic drugs using female rats with exogenous testosterone exposure. METHODS Female rats were injected with testosterone (100 μg/animal) or vehicle on postnatal days 1 to 6. On postnatal day 7, the animals were randomized to either isoflurane exposure or sham. Spatial memory was assessed with the Barnes maze starting on postnatal day 41. Western blots were run from testosterone treated postnatal day 7 animals to measure levels of chloride cotransporters sodium-potassium-chloride symporter (NKCC1) and chloride-potassium symporter 5 (KCC2). RESULTS Exogenous testosterone modulated isoflurane anesthetic neurotoxicity in female rats based on poor performance in the probe trial of the Barnes Maze. By contrast, females with vehicle and isoflurane exposure were able to differentiate the goal position. These behavioral differences corresponded to differences in the protein levels of NKCC1 and KCC2 after exogenous testosterone exposure, with NKCC1 increasing ( P <0.001) and KCC2 decreasing ( P =0.003) relative to female controls. CONCLUSIONS The expression of chloride cotransporters, NKCC1 and KCC2, is altered by testosterone in female rats and corresponds to a cognitive deficit after isoflurane exposure. This confirms the role of androgens in perinatal anesthetic neurotoxicity and supports our hypothesis that the developing GABAergic system plays a critical role in the underlying mechanism.
Collapse
Affiliation(s)
- Gregory A Chinn
- University of California, San Francisco, Department of Anesthesia and Perioperative Care, San Francisco, CA
| | - Katrina Duong
- University of California, San Francisco, Department of Anesthesia and Perioperative Care, San Francisco, CA
| | - Tal R Horovitz
- University of California, San Francisco, Department of Anesthesia and Perioperative Care, San Francisco, CA
| | - Jennifer M Sasaki Russell
- University of California, San Francisco, Department of Anesthesia and Perioperative Care, San Francisco, CA
| | - Jeffrey W Sall
- University of California, San Francisco, Department of Anesthesia and Perioperative Care, San Francisco, CA
| |
Collapse
|
21
|
Urien L, Cohen S, Howard S, Yakimov A, Nordlicht R, Bauer EP. Aversive Contexts Reduce Activity in the Ventral Subiculum- BNST Pathway. Neuroscience 2022; 496:129-140. [PMID: 35724771 PMCID: PMC9329270 DOI: 10.1016/j.neuroscience.2022.06.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 06/08/2022] [Accepted: 06/12/2022] [Indexed: 01/25/2023]
Abstract
Many anxiety disorders can be characterized by abnormalities in detecting and learning about threats, and the inability to reduce fear responses in non-threatening environments. PTSD may be the most representative of context processing pathology, as intrusive memories are experienced in "safe" contexts. The ventral subiculum (vSUB), the main output of the ventral hippocampus, encodes environmental cues and is critical for context processing. The bed nucleus of the stria terminalis (BNST) contributes to anxiety-like behaviors as well as context fear conditioning. Given the important roles of the BNST and the vSUB in these anxiety and fear-related behaviors, and the anatomical connections between the two brain regions, the major aims of this study were to characterize the anatomy and function of the vSUB-BNST pathway. First, using the retrograde tracer cholera toxin, we mapped the topographical arrangement of the vSUB-BNST pathway. Dual retrograde tracing experiments revealed neurons projecting to the BNST and those projecting to the basolateral amygdala are distinct populations. Second, we assessed whether activity in this pathway, as indexed by FOS immunohistochemistry, was modulated by context fear conditioning. Our data reveal less activation of the vSUB-BNST pathway in both males and females in aversive contexts and the greatest activation when animals explored a neutral familiar context. In addition, the vSUB of females contained fewer GABAergic neurons compared to males. These findings suggest that the vSUB-BNST pathway is involved in eliciting appropriate responses to contexts.
Collapse
Affiliation(s)
- Louise Urien
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Stacey Cohen
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Sophia Howard
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Alexandrina Yakimov
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Rachel Nordlicht
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States
| | - Elizabeth P Bauer
- Departments of Biology and Neuroscience & Behavior, Barnard College of Columbia University, 3009 Broadway, New York, NY 10027, United States.
| |
Collapse
|
22
|
Gilfarb RA, Leuner B. GABA System Modifications During Periods of Hormonal Flux Across the Female Lifespan. Front Behav Neurosci 2022; 16:802530. [PMID: 35783228 PMCID: PMC9245048 DOI: 10.3389/fnbeh.2022.802530] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/21/2022] [Indexed: 01/10/2023] Open
Abstract
The female lifespan is marked by periods of dramatic hormonal fluctuation. Changes in the ovarian hormones estradiol and progesterone, in addition to the progesterone metabolite allopregnanolone, are among the most significant and have been shown to have widespread effects on the brain. This review summarizes current understanding of alterations that occur within the GABA system during the major hormonal transition periods of puberty, the ovarian cycle, pregnancy and the postpartum period, as well as reproductive aging. The functional impacts of altered inhibitory activity during these times are also discussed. Lastly, avenues for future research are identified, which, if pursued, can broaden understanding of the GABA system in the female brain and potentially lead to better treatments for women experiencing changes in brain function at each of these hormonal transition periods.
Collapse
Affiliation(s)
- Rachel A. Gilfarb
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, United States
| | - Benedetta Leuner
- Department of Psychology, The Ohio State University, Columbus, OH, United States
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States
- *Correspondence: Benedetta Leuner,
| |
Collapse
|
23
|
Maleninska K, Janikova M, Radostova D, Vojtechova I, Petrasek T, Kirdajova D, Anderova M, Svoboda J, Stuchlik A. Selective deficits in attentional set-shifting in mice induced by maternal immune activation with poly(I:C). Behav Brain Res 2022; 419:113678. [PMID: 34838932 DOI: 10.1016/j.bbr.2021.113678] [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: 09/10/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 11/16/2022]
Abstract
Maternal immune activation has been identified as a significant risk factor for schizophrenia. Using rodent models, past work has demonstrated various behavioral and brain impairments in offspring after immune-activating events. We applied 5 mg/kg of poly(I:C) on gestation day 9 to pregnant mouse dams, whose offspring were then stressed during puberty. We show impairments in attentional set-shifting in a T-maze, and a decreased number of parvalbumin-positive interneurons in the hippocampus as a result of peripubertal stress specifically in females.
Collapse
Affiliation(s)
- Kristyna Maleninska
- Laboratory of the Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 14220, Czech Republic; Faculty of Science, Charles University, Prague, Czech Republic; National Institute of Mental Health, Topolova 748, Klecany, Czech Republic
| | - Martina Janikova
- Laboratory of the Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 14220, Czech Republic; First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Dominika Radostova
- Laboratory of the Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 14220, Czech Republic
| | - Iveta Vojtechova
- Laboratory of the Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 14220, Czech Republic; National Institute of Mental Health, Topolova 748, Klecany, Czech Republic
| | - Tomas Petrasek
- Laboratory of the Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 14220, Czech Republic; National Institute of Mental Health, Topolova 748, Klecany, Czech Republic
| | - Denisa Kirdajova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, Prague 14220, Czech Republic
| | - Miroslava Anderova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, Prague 14220, Czech Republic
| | - Jan Svoboda
- Laboratory of the Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 14220, Czech Republic
| | - Ales Stuchlik
- Laboratory of the Neurophysiology of Memory, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 14220, Czech Republic.
| |
Collapse
|
24
|
Reddy V, McCarthy M, Raval AP. Xenoestrogens impact brain estrogen receptor signaling during the female lifespan: A precursor to neurological disease? Neurobiol Dis 2021; 163:105596. [PMID: 34942334 DOI: 10.1016/j.nbd.2021.105596] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/08/2021] [Accepted: 12/18/2021] [Indexed: 02/07/2023] Open
Abstract
Xenoestrogens, foreign synthetic chemicals mimicking estrogens, are lurking in our surroundings. Climate change may alter their toxicity and bioavailability. Since xenoestrogens have extremely high lipid solubility and are structurally similar to natural endogenous estrogens, they can bind to estrogen receptors (ERs) -alpha (ER-α) and -beta (ER-β). Scientific evidence accumulated over the past decades have suggested that natural 17β-estradiol (E2; a potent estrogen), via activation of its receptors, plays a pivotal role in regulation of brain development, differentiation, metabolism, synaptic plasticity, neuroprotection, cognition, anxiety, body temperature, feeding and sexual behavior. In the brain, ER-β is predominantly expressed in the various regions, including cerebral cortex and hippocampus, that have been shown to play a key role in cognition. Therefore, disturbances in function of ER-β mediated E2 signaling by xenoestrogens can lead to deleterious effects that potentiate a variety of neurological diseases starting from prenatal to post-menopause in women. The goal of this review is to identify the possible neurological effects of xenoestrogens that can alter estrogen receptor-mediated signaling in the brain during different stages of the female lifespan.
Collapse
Affiliation(s)
- Varun Reddy
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA; Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Micheline McCarthy
- Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA; Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, 33136, USA.
| |
Collapse
|
25
|
Woodward EM, Coutellier L. Age- and sex-specific effects of stress on parvalbumin interneurons in preclinical models: Relevance to sex differences in clinical neuropsychiatric and neurodevelopmental disorders. Neurosci Biobehav Rev 2021; 131:1228-1242. [PMID: 34718048 PMCID: PMC8642301 DOI: 10.1016/j.neubiorev.2021.10.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/06/2021] [Accepted: 10/23/2021] [Indexed: 01/06/2023]
Abstract
Stress is a major risk factor for neurodevelopmental and neuropsychiatric disorders, with the capacity to impact susceptibility to disease as well as long-term neurobiological and behavioral outcomes. Parvalbumin (PV) interneurons, the most prominent subtype of GABAergic interneurons in the cortex, are uniquely responsive to stress due to their protracted development throughout the highly plastic neonatal period and into puberty and adolescence. Additionally, PV + interneurons appear to respond to stress in a sex-specific manner. This review aims to discuss existing preclinical studies that support our overall hypothesis that the sex-and age-specific impacts of stress on PV + interneurons contribute to differences in individual vulnerability to stress across the lifespan, particularly in regard to sex differences in the diagnostic rate of neurodevelopmental and neuropsychiatric diseases in clinical populations. We also emphasize the importance of studying sex as a biological variable to fully understand the mechanistic and behavioral differences between males and females in models of neuropsychiatric disease.
Collapse
Affiliation(s)
- Emma M Woodward
- Department of Neuroscience, Ohio State University, 255 Institute for Behavioral Medicine Research Building, 460 Medical Center Drive, Columbus, OH, 43210, United States
| | - Laurence Coutellier
- Department of Neuroscience, Ohio State University, 255 Institute for Behavioral Medicine Research Building, 460 Medical Center Drive, Columbus, OH, 43210, United States; Department of Psychology, Ohio State University, 53 Psychology Building, 1835 Neil Avenue, Columbus, OH, 43210, United States.
| |
Collapse
|
26
|
Ellis SN, Honeycutt JA. Sex Differences in Affective Dysfunction and Alterations in Parvalbumin in Rodent Models of Early Life Adversity. Front Behav Neurosci 2021; 15:741454. [PMID: 34803622 PMCID: PMC8600234 DOI: 10.3389/fnbeh.2021.741454] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/13/2021] [Indexed: 01/08/2023] Open
Abstract
The early life environment markedly influences brain and behavioral development, with adverse experiences associated with increased risk of anxiety and depressive phenotypes, particularly in females. Indeed, early life adversity (ELA) in humans (i.e., caregiver deprivation, maltreatment) and rodents (i.e., maternal separation, resource scarcity) is associated with sex-specific emergence of anxious and depressive behaviors. Although these disorders show clear sex differences in humans, little attention has been paid toward evaluating sex as a biological variable in models of affective dysfunction; however, recent rodent work suggests sex-specific effects. Two widely used rodent models of ELA approximate caregiver deprivation (i.e., maternal separation) and resource scarcity (i.e., limited bedding). While these approaches model aspects of ELA experienced in humans, they span different portions of the pre-weaning developmental period and may therefore differentially contribute to underlying mechanistic risk. This is borne out in the literature, where evidence suggests differences in trajectories of behavior depending on the type of ELA and/or sex; however, the neural underpinning of these differences is not well understood. Because anxiety and depression are thought to involve dysregulation in the balance of excitatory and inhibitory signaling in ELA-vulnerable brain regions (e.g., prefrontal cortex, amygdala, hippocampus), outcomes are likely driven by alterations in local and/or circuit-specific inhibitory activity. The most abundant GABAergic subtypes in the brain, accounting for approximately 40% of inhibitory neurons, contain the calcium-binding protein Parvalbumin (PV). As PV-expressing neurons have perisomatic and proximal dendritic targets on pyramidal neurons, they are well-positioned to regulate excitatory/inhibitory balance. Recent evidence suggests that PV outcomes following ELA are sex, age, and region-specific and may be influenced by the type and timing of ELA. Here, we suggest the possibility of a combined role of PV and sex hormones driving differences in behavioral outcomes associated with affective dysfunction following ELA. This review evaluates the literature across models of ELA to characterize neural (PV) and behavioral (anxiety- and depressive-like) outcomes as a function of sex and age. Additionally, we detail a putative mechanistic role of PV on ELA-related outcomes and discuss evidence suggesting hormone influences on PV expression/function which may help to explain sex differences in ELA outcomes.
Collapse
Affiliation(s)
- Seneca N Ellis
- Program in Neuroscience, Bowdoin College, Brunswick, ME, United States
| | - Jennifer A Honeycutt
- Program in Neuroscience, Bowdoin College, Brunswick, ME, United States.,Department of Psychology, Bowdoin College, Brunswick, ME, United States
| |
Collapse
|
27
|
Lum JS, Bird KM, Wilkie J, Millard SJ, Pallimulla S, Newell KA, Wright IM. Prenatal methadone exposure impairs adolescent cognition and GABAergic neurodevelopment in a novel rat model of maternal methadone treatment. Prog Neuropsychopharmacol Biol Psychiatry 2021; 110:110281. [PMID: 33571606 DOI: 10.1016/j.pnpbp.2021.110281] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 01/21/2021] [Accepted: 02/05/2021] [Indexed: 10/22/2022]
Abstract
Methadone maintenance treatment (MMT) is the most common treatment for opioid-dependent pregnant women worldwide. Despite its widespread use, MMT is associated with a variety of adverse neurodevelopmental outcomes in exposed offspring, particularly cognitive impairments. The neurobiological abnormalities underlying these cognitive impairments are, however, poorly understood. This is, in part, due to a lack of animal models that represents the standard of care that methadone is administered in the clinic, with inconsistencies in the timing, doses and durations of treatment. Here we describe the characterisation of a clinically relevant rat model of MMT in which the long-term behavioural and neurobiological effects of prenatal methadone exposure can be assessed in adolescent offspring. Female Sprague-Dawley rats were treated orally with an ascending methadone dosage schedule (5, 10, 15, 20, 25 and 30 mg/kg/day), self-administered in drinking water prior to conception, throughout gestation and lactation. Pregnancy success, maternal gestational weight gain, litter survival and size were not significantly altered in methadone-exposed animals. Methadone-exposed offspring body and brain weights were significantly lower at birth. Novel object recognition tests performed at adolescence revealed methadone-exposed offspring had impaired recognition memory. Furthermore, the rewarded T-maze alternation task demonstrated that methadone-exposed female, but not male, offspring also exhibit working memory and learning deficits. Immunoblots of the adolescent prefrontal cortex and hippocampus showed methadone-exposed offspring displayed reduced levels of mature BDNF, in addition to the GABAergic proteins, GAD67 and parvalbumin, in a sex- and brain region-specific fashion. This rat model closely emulates the clinical scenario in which methadone is administered to opioid-dependent pregnant woman and provides evidence MMT can cause cognitive impairments in adolescent offspring that may be underlined by perturbed neurodevelopment of the GABAergic system.
Collapse
Affiliation(s)
- Jeremy S Lum
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; School of Medicine, University of Wollongong University of Wollongong, Wollongong, NSW 2522, Australia; Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia.
| | - Katrina M Bird
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; School of Medicine, University of Wollongong University of Wollongong, Wollongong, NSW 2522, Australia; Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Jennifer Wilkie
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; School of Medicine, University of Wollongong University of Wollongong, Wollongong, NSW 2522, Australia
| | - Samuel J Millard
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; School of Medicine, University of Wollongong University of Wollongong, Wollongong, NSW 2522, Australia
| | - Sachie Pallimulla
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; School of Medicine, University of Wollongong University of Wollongong, Wollongong, NSW 2522, Australia
| | - Kelly A Newell
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; School of Medicine, University of Wollongong University of Wollongong, Wollongong, NSW 2522, Australia; Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Ian M Wright
- Illawarra Health and Medical Research Institute, Wollongong, NSW 2522, Australia; School of Medicine, University of Wollongong University of Wollongong, Wollongong, NSW 2522, Australia; University of Queensland Centre for Clinical Research, University of Queensland, Herston, QLD 4029, Australia; College of Medicine and Dentistry, James Cook University, Cairns, QLD 4870, Australia
| |
Collapse
|
28
|
Trova S, Bovetti S, Bonzano S, De Marchis S, Peretto P. Sex Steroids and the Shaping of the Peripubertal Brain: The Sexual-Dimorphic Set-Up of Adult Neurogenesis. Int J Mol Sci 2021; 22:ijms22157984. [PMID: 34360747 PMCID: PMC8347822 DOI: 10.3390/ijms22157984] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 12/29/2022] Open
Abstract
Steroid hormones represent an amazing class of molecules that play pleiotropic roles in vertebrates. In mammals, during postnatal development, sex steroids significantly influence the organization of sexually dimorphic neural circuits underlying behaviors critical for survival, such as the reproductive one. During the last decades, multiple studies have shown that many cortical and subcortical brain regions undergo sex steroid-dependent structural organization around puberty, a critical stage of life characterized by high sensitivity to external stimuli and a profound structural and functional remodeling of the organism. Here, we first give an overview of current data on how sex steroids shape the peripubertal brain by regulating neuroplasticity mechanisms. Then, we focus on adult neurogenesis, a striking form of persistent structural plasticity involved in the control of social behaviors and regulated by a fine-tuned integration of external and internal cues. We discuss recent data supporting that the sex steroid-dependent peripubertal organization of neural circuits involves a sexually dimorphic set-up of adult neurogenesis that in turn could be relevant for sex-specific reproductive behaviors.
Collapse
Affiliation(s)
- Sara Trova
- Department of Life Sciences and Systems Biology (DBIOS), University of Torino, 10123 Turin, Italy; (S.T.); (S.B.); (S.B.); (S.D.M.)
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, 10043 Turin, Italy
| | - Serena Bovetti
- Department of Life Sciences and Systems Biology (DBIOS), University of Torino, 10123 Turin, Italy; (S.T.); (S.B.); (S.B.); (S.D.M.)
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, 10043 Turin, Italy
| | - Sara Bonzano
- Department of Life Sciences and Systems Biology (DBIOS), University of Torino, 10123 Turin, Italy; (S.T.); (S.B.); (S.B.); (S.D.M.)
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, 10043 Turin, Italy
| | - Silvia De Marchis
- Department of Life Sciences and Systems Biology (DBIOS), University of Torino, 10123 Turin, Italy; (S.T.); (S.B.); (S.B.); (S.D.M.)
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, 10043 Turin, Italy
| | - Paolo Peretto
- Department of Life Sciences and Systems Biology (DBIOS), University of Torino, 10123 Turin, Italy; (S.T.); (S.B.); (S.B.); (S.D.M.)
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, 10043 Turin, Italy
- Correspondence:
| |
Collapse
|
29
|
Castagna PJ, Crowley MJ. Relationship between Puberty and Inhibitory Control: Computational Modeling of the Drift-diffusion Process. Dev Neuropsychol 2021; 46:360-380. [PMID: 34283678 DOI: 10.1080/87565641.2021.1952206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Previous work relies largely on the simple reaction time measures in inhibitory control tasks. The goal of the current study was to provide a better understanding the relationship between puberty, sex, and inhibitory control utilizing and contrasting two popular drift diffusion models. A sample of 103 adolescents (Mage = 14.49, SD = 1.69) self-reported their pubertal development and completed a flanker task. Utilizing Bayesian regressions, we found that the interaction between puberty and sex were significant predictors of the A/B parameter, conceptualized as the amount of information considered for a decision during the task.
Collapse
Affiliation(s)
- Peter J Castagna
- Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA
| | - Michael J Crowley
- Yale Child Study Center, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
30
|
Bueno-Fernandez C, Perez-Rando M, Alcaide J, Coviello S, Sandi C, Castillo-Gómez E, Nacher J. Long term effects of peripubertal stress on excitatory and inhibitory circuits in the prefrontal cortex of male and female mice. Neurobiol Stress 2021; 14:100322. [PMID: 33869684 PMCID: PMC8045050 DOI: 10.1016/j.ynstr.2021.100322] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/22/2021] [Accepted: 03/24/2021] [Indexed: 12/01/2022] Open
Abstract
The impact of stressful events is especially important during early life, because certain cortical regions, especially the prefrontal cortex (PFC), are still developing. Consequently, aversive experiences that occur during the peripubertal period can cause long-term alterations in neural connectivity, physiology and related behaviors. Although sex influences the stress response and women are more likely to develop stress-related psychiatric disorders, knowledge about the effects of stress on females is still limited. In order to analyze the long-term effects of peripubertal stress (PPS) on the excitatory and inhibitory circuitry of the adult PFC, and whether these effects are sex-dependent, we applied an unpredictable chronic PPS protocol based on psychogenic stressors. Using two strains of transgenic mice with specific fluorescent cell reporters, we studied male and diestrus females to know how PPS affects the structure and connectivity of parvalbumin expressing (PV+) interneurons and pyramidal neurons. We also studied the expression of molecules related to excitatory and inhibitory neurotransmission, as well as alterations in the expression of plasticity-related molecules. The structure of pyramidal neurons was differentially affected by PPS in male and female mice: while the former had a decreased dendritic spine density, the latter displayed an increase in this parameter. PPS affected the density of puncta expressing excitatory and inhibitory synaptic markers exclusively in the female mPFC. Similarly, only in female mice we observed an increased complexity of the dendritic tree of PV+ neurons. Regarding the perisomatic innervation on pyramidal and PV + neurons by basket cells, we found a significant increase in the density of puncta in stressed animals, with interesting differences between the sexes and the type of basket cell analyzed. Finally, the PPS protocol also altered the total number of somata expressing the polysialylated form of the neural cell adhesion molecule (PSA-NCAM) when we analyzed both sexes together. These results highlight the strong programming effects of aversive experiences during early life for the establishment of cortical circuitry and the special impact of these stressful events on females.
Collapse
Affiliation(s)
- Clara Bueno-Fernandez
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain
| | - Marta Perez-Rando
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain
| | - Julia Alcaide
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain
| | - Simona Coviello
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain
| | - Carmen Sandi
- Department of Life Sciences, Laboratory of Behavioral Genetics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Esther Castillo-Gómez
- Department of Medicine, School of Medical Sciences, Universitat Jaume I, Valencia, Spain.,Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain
| | - Juan Nacher
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain.,Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain.,Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain
| |
Collapse
|
31
|
Que L, Lukacsovich D, Luo W, Földy C. Transcriptional and morphological profiling of parvalbumin interneuron subpopulations in the mouse hippocampus. Nat Commun 2021; 12:108. [PMID: 33398060 PMCID: PMC7782706 DOI: 10.1038/s41467-020-20328-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 11/27/2020] [Indexed: 12/21/2022] Open
Abstract
The diversity reflected by >100 different neural cell types fundamentally contributes to brain function and a central idea is that neuronal identity can be inferred from genetic information. Recent large-scale transcriptomic assays seem to confirm this hypothesis, but a lack of morphological information has limited the identification of several known cell types. In this study, we used single-cell RNA-seq in morphologically identified parvalbumin interneurons (PV-INs), and studied their transcriptomic states in the morphological, physiological, and developmental domains. Overall, we find high transcriptomic similarity among PV-INs, with few genes showing divergent expression between morphologically different types. Furthermore, PV-INs show a uniform synaptic cell adhesion molecule (CAM) profile, suggesting that CAM expression in mature PV cells does not reflect wiring specificity after development. Together, our results suggest that while PV-INs differ in anatomy and in vivo activity, their continuous transcriptomic and homogenous biophysical landscapes are not predictive of these distinct identities.
Collapse
Affiliation(s)
- Lin Que
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich, Switzerland
| | - David Lukacsovich
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich, Switzerland
| | - Wenshu Luo
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich, Switzerland
| | - Csaba Földy
- Laboratory of Neural Connectivity, Brain Research Institute, Faculties of Medicine and Science, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
32
|
Rurak GM, Woodside B, Aguilar-Valles A, Salmaso N. Astroglial cells as neuroendocrine targets in forebrain development: Implications for sex differences in psychiatric disease. Front Neuroendocrinol 2021; 60:100897. [PMID: 33359797 DOI: 10.1016/j.yfrne.2020.100897] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/05/2020] [Accepted: 12/15/2020] [Indexed: 12/23/2022]
Abstract
Astroglial cells are the most abundant cell type in the mammalian brain. They are implicated in almost every aspect of brain physiology, including maintaining homeostasis, building and maintaining the blood brain barrier, and the development and maturation of neuronal networks. Critically, astroglia also express receptors for gonadal sex hormones, respond rapidly to gonadal hormones, and are able to synthesize hormones. Thus, they are positioned to guide and mediate sexual differentiation of the brain, particularly neuronal networks in typical and pathological conditions. In this review, we describe astroglial involvement in the organization and development of the brain, and consider known sex differences in astroglial responses to understand how astroglial cell-mediated organization may play a role in forebrain sexual dimorphisms in human populations. Finally, we consider how sexually dimorphic astroglial responses and functions in development may lead to sex differences in vulnerability for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Gareth M Rurak
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Barbara Woodside
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada; Concordia University, Montreal, Quebec, Canada
| | | | - Natalina Salmaso
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada.
| |
Collapse
|
33
|
Laube C, Fuhrmann D. Is early good or bad? Early puberty onset and its consequences for learning. Curr Opin Behav Sci 2020. [DOI: 10.1016/j.cobeha.2020.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
34
|
Hill RA, Kouremenos K, Tull D, Maggi A, Schroeder A, Gibbons A, Kulkarni J, Sundram S, Du X. Bazedoxifene - a promising brain active SERM that crosses the blood brain barrier and enhances spatial memory. Psychoneuroendocrinology 2020; 121:104830. [PMID: 32858306 DOI: 10.1016/j.psyneuen.2020.104830] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 08/11/2020] [Indexed: 12/21/2022]
Abstract
Over 20 years of accumulated evidence has shown that the major female sex hormone 17β-estradiol can enhance cognitive functioning. However, the utility of estradiol as a therapeutic cognitive enhancer is hindered by its unwanted peripheral effects (carcinogenic). Selective estrogen receptor modulators (SERMs) avoid the unwanted effects of estradiol by acting as estrogen receptor antagonists in some tissues such as breast and uterus, but as agonists in others such as bone, and are currently used for the treatment of osteoporosis. However, understanding of their actions in the brain are limited. The third generation SERM bazedoxifene has recently been FDA approved for clinical use with an improved biosafety profile. However, whether bazedoxifene can enter the brain and enhance cognition is unknown. Using mice, the current study aimed to explore if bazedoxifene can 1) cross the blood-brain barrier, 2) rescue ovariectomy-induced hippocampal-dependent spatial memory deficit, and 3) activate neural estrogen response element (ERE)-dependent gene transcription. Using liquid chromatography-mass spectrometry (LC-MS), we firstly demonstrate that a peripheral injection of bazedoxifene can enter the brain. Secondly, we show that an acute intraperitoneal injection of bazedoxifene can rescue ovariectomy-induced spatial memory deficits. And finally, using the ERE-luciferase reporter mouse, we show in vivo that bazedoxifene can activate the ERE in the brain. The evidence shown here suggest bazedoxifene could be a viable cognitive enhancer with promising clinical applicability.
Collapse
Affiliation(s)
- R A Hill
- Department of Psychiatry, Monash University, Clayton, VIC, 3168, Australia; Florey Institute for Neuroscience and Mental Health, Parkville, VIC, 3052, Australia.
| | - K Kouremenos
- Metabolomics Australia, Bio21 Molecular Science & Biotechnology Institute, Parkville, VIC, 3052, Australia
| | - D Tull
- Metabolomics Australia, Bio21 Molecular Science & Biotechnology Institute, Parkville, VIC, 3052, Australia
| | - A Maggi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, 20133, Italy
| | - A Schroeder
- Department of Psychiatry, Monash University, Clayton, VIC, 3168, Australia
| | - A Gibbons
- Department of Psychiatry, Monash University, Clayton, VIC, 3168, Australia
| | - J Kulkarni
- Monash Alfred Psychiatry Research Centre, Monash University, St Kilda, VIC, 3004, Australia
| | - S Sundram
- Department of Psychiatry, Monash University, Clayton, VIC, 3168, Australia
| | - X Du
- Department of Psychiatry, Monash University, Clayton, VIC, 3168, Australia; Florey Institute for Neuroscience and Mental Health, Parkville, VIC, 3052, Australia
| |
Collapse
|
35
|
Notaras M, van den Buuse M. Neurobiology of BDNF in fear memory, sensitivity to stress, and stress-related disorders. Mol Psychiatry 2020; 25:2251-2274. [PMID: 31900428 DOI: 10.1038/s41380-019-0639-2] [Citation(s) in RCA: 228] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 12/01/2019] [Accepted: 12/12/2019] [Indexed: 01/17/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is widely accepted for its involvement in resilience and antidepressant drug action, is a common genetic locus of risk for mental illnesses, and remains one of the most prominently studied molecules within psychiatry. Stress, which arguably remains the "lowest common denominator" risk factor for several mental illnesses, targets BDNF in disease-implicated brain regions and circuits. Altered stress-related responses have also been observed in animal models of BDNF deficiency in vivo, and BDNF is a common downstream intermediary for environmental factors that potentiate anxiety- and depressive-like behavior. However, BDNF's broad functionality has manifested a heterogeneous literature; likely reflecting that BDNF plays a hitherto under-recognized multifactorial role as both a regulator and target of stress hormone signaling within the brain. The role of BDNF in vulnerability to stress and stress-related disorders, such as posttraumatic stress disorder (PTSD), is a prominent example where inconsistent effects have emerged across numerous models, labs, and disciplines. In the current review we provide a contemporary update on the neurobiology of BDNF including new data from the behavioral neuroscience and neuropsychiatry literature on fear memory consolidation and extinction, stress, and PTSD. First we present an overview of recent advances in knowledge on the role of BDNF within the fear circuitry, as well as address mounting evidence whereby stress hormones interact with endogenous BDNF-TrkB signaling to alter brain homeostasis. Glucocorticoid signaling also acutely recruits BDNF to enhance the expression of fear memory. We then include observations that the functional common BDNF Val66Met polymorphism modulates stress susceptibility as well as stress-related and stress-inducible neuropsychiatric endophenotypes in both man and mouse. We conclude by proposing a BDNF stress-sensitivity hypothesis, which posits that disruption of endogenous BDNF activity by common factors (such as the BDNF Val66Met variant) potentiates sensitivity to stress and, by extension, vulnerability to stress-inducible illnesses. Thus, BDNF may induce plasticity to deleteriously promote the encoding of fear and trauma but, conversely, also enable adaptive plasticity during extinction learning to suppress PTSD-like fear responses. Ergo regulators of BDNF availability, such as the Val66Met polymorphism, may orchestrate sensitivity to stress, trauma, and risk of stress-induced disorders such as PTSD. Given an increasing interest in personalized psychiatry and clinically complex cases, this model provides a framework from which to experimentally disentangle the causal actions of BDNF in stress responses, which likely interact to potentiate, produce, and impair treatment of, stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Michael Notaras
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, Cornell University, New York, NY, USA.
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia. .,College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia. .,Department of Pharmacology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
36
|
Hill RA, Grech AM, Notaras MJ, Sepulveda M, van den Buuse M. Brain-Derived Neurotrophic Factor Val66Met polymorphism interacts with adolescent stress to alter hippocampal interneuron density and dendritic morphology in mice. Neurobiol Stress 2020; 13:100253. [PMID: 33344708 PMCID: PMC7739172 DOI: 10.1016/j.ynstr.2020.100253] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/03/2020] [Accepted: 09/26/2020] [Indexed: 01/06/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) plays essential roles in GABAergic interneuron development. The common BDNF val66met polymorphism, leads to decreased activity-dependent release of BDNF. The current study used a humanized mouse model of the BDNF val66met polymorphism to determine how reduced activity-dependent release of BDNF, both on its own, and in combination with chronic adolescent stress hormone, impact hippocampal GABAergic interneuron cell density and dendrite morphology. Male and female Val/Val and Met/Met mice were exposed to corticosterone (CORT) or placebo in their drinking water from weeks 6-8, before brains were perfuse-fixed at 15 weeks. Cell density and dendrite morphology of immunofluorescent labelled inhibitory interneurons; somatostatin, parvalbumin and calretinin in the CA1, and 3 and dentate gyrus (DG) across the dorsal (DHP) and ventral hippocampus (VHP) were assessed by confocal z-stack imaging, and IMARIS dendritic mapping software. Mice with the Met/Met genotype showed significantly lower somatostatin cell density compared to Val/Val controls in the DHP, and altered somatostatin interneuron dendrite morphology including branch depth, and spine density. Parvalbumin-positive interneurons were unchanged between genotype groups, however BDNF val66met genotype influenced the dendritic volume, branch level and spine density of parvalbumin interneurons differentially across hippocampal subregions. Contrary to this, no such effects were observed for calretinin-positive interneurons. Adolescent exposure to CORT treatment also significantly altered somatostatin and parvalbumin dendrite branch level and the combined effect of Met/Met genotype and CORT treatment significantly reduced somatostatin and parvalbumin dendrite spine density. In sum, the BDNFVal66Met polymorphism significantly alters somatostatin and parvalbumin-positive interneuron cell development and dendrite morphology. Additionally, we also report a compounding effect of the Met/Met genotype and chronic adolescent CORT treatment on dendrite spine density, indicating that adolescence is a sensitive period of risk for Val66Met polymorphism carriers.
Collapse
Affiliation(s)
- Rachel Anne Hill
- Department of Psychiatry, School of Clinical Sciences, Monash University, Monash Medical Centre, Clayton, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- Corresponding author. Department of Psychiatry, School of Clinical Sciences at Monash Health, Monash University Level 3, Monash Medical Centre 27Wright St Clayton VIC 3168 Australia, .
| | - Adrienne Mary Grech
- Department of Psychiatry, School of Clinical Sciences, Monash University, Monash Medical Centre, Clayton, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Michael J. Notaras
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- Weill Cornell Medical College of Cornell University, Centre for Neurogenetics, Brain & Mind Research Institute, New York City, New York, USA
| | - Mauricio Sepulveda
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
- Department of Pharmacology, University of Melbourne, VIC, Australia
- The College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
| |
Collapse
|
37
|
Torromino G, Maggi A, De Leonibus E. Estrogen-dependent hippocampal wiring as a risk factor for age-related dementia in women. Prog Neurobiol 2020; 197:101895. [PMID: 32781107 DOI: 10.1016/j.pneurobio.2020.101895] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/22/2020] [Accepted: 08/03/2020] [Indexed: 02/08/2023]
Abstract
Women are more prone than men to develop age-related dementia, such as Alzheimer's disease (AD). This has been linked to the marked decrease in circulating estrogens during menopause. This review proposes to change this perspective and consider women's vulnerability to developing AD as a consequence of sex differences in the neurobiology of memory, focusing on the hippocampus. The hippocampus of cognitively impaired subjects tends to shrink with age; however, in many cases, this can be prevented by exercise or cognitive training, suggesting that if you do not use the hippocampus you lose it. We will review the developmental trajectory of sex steroids-regulated differences on the hippocampus, proposing that the overall shaping action of sex-steroids results in a lower usage of the hippocampus in females, which in turn makes them more vulnerable to the effects of ageing, the "network fragility hypothesis". To explain why women rely less on hippocampus-dependent strategies, we propose a "computational hypothesis" that is based on experimental evidence suggesting that the direct effects of estrogens on hippocampal synaptic and structural plasticity during the estrous-cycle confers instability to the memory-dependent hippocampal network. Finally, we propose to counteract AD with training and/or treatments, such as orienteering, which specifically favour the use of the hippocampus.
Collapse
Affiliation(s)
- Giulia Torromino
- Telethon Institute of Genetics and Medicine (TIGEM), Telethon Foundation, Pozzuoli, Naples, Italy; Institute of Biochemistry and Cell Biology (IBBC), National Research Council, Monterotondo, Rome, Italy
| | - Adriana Maggi
- Center of Excellence on Neurodegenerative Diseases, University of Milan, Milan, Italy
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine (TIGEM), Telethon Foundation, Pozzuoli, Naples, Italy; Institute of Biochemistry and Cell Biology (IBBC), National Research Council, Monterotondo, Rome, Italy.
| |
Collapse
|
38
|
Bettio LEB, Thacker JS, Rodgers SP, Brocardo PS, Christie BR, Gil-Mohapel J. Interplay between hormones and exercise on hippocampal plasticity across the lifespan. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165821. [PMID: 32376385 DOI: 10.1016/j.bbadis.2020.165821] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 04/19/2020] [Accepted: 04/25/2020] [Indexed: 12/15/2022]
Abstract
The hippocampus is a brain structure known to play a central role in cognitive function (namely learning and memory) as well as mood regulation and affective behaviors due in part to its ability to undergo structural and functional changes in response to intrinsic and extrinsic stimuli. While structural changes are achieved through modulation of hippocampal neurogenesis as well as alterations in dendritic morphology and spine remodeling, functional (i.e., synaptic) changes can be noted through the strengthening (i.e., long-term potentiation) or weakening (i.e., long-term depression) of the synapses. While age, hormone homeostasis, and levels of physical activity are some of the factors known to module these forms of hippocampal plasticity, the exact mechanisms through which these factors interact with each other at a given moment in time are not completely understood. It is well known that hormonal levels vary throughout the lifespan of an individual and it is also known that physical exercise can impact hormonal homeostasis. Thus, it is reasonable to speculate that hormone modulation might be one of the various mechanisms through which physical exercise differently impacts hippocampal plasticity throughout distinct periods of an individual's life. The present review summarizes the potential relationship between physical exercise and different types of hormones (namely sex, metabolic, and stress hormones) and how this relationship may mediate the effects of physical activity during three distinct life periods, adolescence, adulthood, and senescence. Overall, the vast majority of studies support a beneficial role of exercise in maintaining hippocampal hormonal levels and consequently, hippocampal plasticity, cognition, and mood regulation.
Collapse
Affiliation(s)
- Luis E B Bettio
- Division of Medical Sciences and Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
| | - Jonathan S Thacker
- Division of Medical Sciences and Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
| | - Shaefali P Rodgers
- Developmental, Cognitive & Behavioral Neuroscience Program, Department of Psychology, Texas Institute for Measurement, Evaluation, and Statistics, University of Houston, TX, USA
| | - Patricia S Brocardo
- Department of Morphological Sciences, Centre of Biological Sciences, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Brian R Christie
- Division of Medical Sciences and Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada; Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC, Canada
| | - Joana Gil-Mohapel
- Division of Medical Sciences and Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada; Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC, Canada.
| |
Collapse
|
39
|
Shen H, Kenney L, Smith SS. Increased Dendritic Branching of and Reduced δ-GABA A Receptor Expression on Parvalbumin-Positive Interneurons Increase Inhibitory Currents and Reduce Synaptic Plasticity at Puberty in Female Mouse CA1 Hippocampus. Front Cell Neurosci 2020; 14:203. [PMID: 32733208 PMCID: PMC7363981 DOI: 10.3389/fncel.2020.00203] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/10/2020] [Indexed: 12/04/2022] Open
Abstract
Parvalbumin positive (PV+) interneurons play a pivotal role in cognition and are known to be regulated developmentally and by ovarian hormones. The onset of puberty represents the end of a period of optimal learning when impairments in synaptic plasticity are observed in the CA1 hippocampus of female mice. Therefore, we tested whether the synaptic inhibitory current generated by PV+ interneurons is increased at puberty and contributes to these deficits in synaptic plasticity. To this end, the spontaneous inhibitory postsynaptic current (sIPSC) was recorded using whole-cell patch-clamp techniques from CA1 pyramidal cells in the hippocampal slice before (PND 28–32) and after the onset of puberty in female mice (~PND 35–44, assessed by vaginal opening). sIPSC frequency and amplitude were significantly increased at puberty, but these measures were reduced by 1 μM DAMGO [1 μM, (D-Ala2, N-MePhe4, Gly-ol)-enkephalin], which silences PV+ activity via μ-opioid receptor targets. At puberty, dendritic branching of PV+ interneurons in GAD67-GFP mice was increased, while expression of the δ subunit of the GABAA receptor (GABAR) on these interneurons decreased. Both frequency and amplitude of sIPSCs were significantly increased in pre-pubertal mice with reduced δ expression, suggesting a possible mechanism. Theta burst induction of long-term potentiation (LTP), an in vitro model of learning, is impaired at puberty but was restored to optimal levels by DAMGO administration, implicating inhibition via PV+ interneurons as one cause. Administration of the neurosteroid/stress steroid THP (30 nM, 3α-OH, 5α-pregnan-20-one) had no effect on sIPSCs. These findings suggest that phasic inhibition generated by PV+ interneurons is increased at puberty when it contributes to impairments in synaptic plasticity. These results may have relevance for the changes in cognitive function reported during early adolescence.
Collapse
Affiliation(s)
- Hui Shen
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, United States.,Research Institute of Neurology, General Hospital, Tianjin Medical University, Heping District, Tianjin, China
| | - Lindsay Kenney
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, United States.,Program in Neural and Behavioral Science, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Sheryl S Smith
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY, United States.,The Robert F. Furchgott Center for Neural and Behavioral Science, SUNY Downstate Medical Center, Brooklyn, NY, United States
| |
Collapse
|
40
|
Laube C, van den Bos W, Fandakova Y. The relationship between pubertal hormones and brain plasticity: Implications for cognitive training in adolescence. Dev Cogn Neurosci 2020; 42:100753. [PMID: 32072931 PMCID: PMC7005587 DOI: 10.1016/j.dcn.2020.100753] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 12/20/2019] [Accepted: 01/07/2020] [Indexed: 12/12/2022] Open
Abstract
Adolescence may mark a sensitive period for the development of higher-order cognition through enhanced plasticity of cortical circuits. At the same time, animal research indicates that pubertal hormones may represent one key mechanism for closing sensitive periods in the associative neocortex, thereby resulting in decreased plasticity of cortical circuits in adolescence. In the present review, we set out to solve some of the existing ambiguity and examine how hormonal changes associated with pubertal onset may modulate plasticity in higher-order cognition during adolescence. We build on existing age-comparative cognitive training studies to explore how the potential for change in neural resources and behavioral repertoire differs across age groups. We review animal and human brain imaging studies, which demonstrate a link between brain development, neurochemical mechanisms of plasticity, and pubertal hormones. Overall, the existent literature indicates that pubertal hormones play a pivotal role in regulating the mechanisms of experience-dependent plasticity during adolescence. However, the extent to which hormonal changes associated with pubertal onset increase or decrease brain plasticity may depend on the specific cognitive domain, the sex, and associated brain networks. We discuss implications for future research and suggest that systematical longitudinal assessments of pubertal change together with cognitive training interventions may be a fruitful way toward a better understanding of adolescent plasticity. As the age of pubertal onset is decreasing across developed societies, this may also have important educational and clinical implications, especially with respect to the effects that earlier puberty has on learning.
Collapse
Affiliation(s)
- Corinna Laube
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany
| | | | - Yana Fandakova
- Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany.
| |
Collapse
|
41
|
Monte AS, da Silva FER, Lima CNDC, Vasconcelos GS, Gomes NS, Miyajima F, Vasconcelos SMM, Gama CS, Seeman MV, de Lucena DF, Macedo DS. Sex influences in the preventive effects of N-acetylcysteine in a two-hit animal model of schizophrenia. J Psychopharmacol 2020; 34:125-136. [PMID: 31556775 DOI: 10.1177/0269881119875979] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Schizophrenia (SCZ) is a neurodevelopmental disorder influenced by patient sex. Mechanisms underlying sex differences in SCZ remain unknown. A two-hit model of SCZ combines the exposure to perinatal infection (first-hit) with peripubertal unpredictable stress (PUS, second-hit). N-acetylcysteine (NAC) has been tested in SCZ because of the involvement of glutathione mechanisms in its neurobiology. AIMS We aim to investigate whether NAC administration to peripubertal rats of both sexes could prevent behavioral and neurochemical changes induced by the two-hit model. METHODS Wistar rats were exposed to polyinosinic:polycytidylic acid (a viral mimetic) or saline on postnatal days (PND) 5-7. On PND30-59 they received saline or NAC 220 mg/kg and between PND40-48 were subjected to PUS or left undisturbed. On PND60 behavioral and oxidative alterations were evaluated in the prefrontal cortex (PFC) and striatum. Mechanisms of hippocampal memory regulation such as immune expression of G protein-coupled estrogen receptor 1 (GPER), α7-nAChR and parvalbumin were also evaluated. RESULTS NAC prevented sensorimotor gating deficits only in females, while it prevented alterations in social interaction, working memory and locomotor activity in both sexes. Again, in rats of both sexes, NAC prevented the following neurochemical alterations: glutathione (GSH) and nitrite levels in the PFC and lipid peroxidation in the PFC and striatum. Striatal oxidative alterations in GSH and nitrite were observed in females and prevented by NAC. Two-hit induced hippocampal alterations in females, namely expression of GPER-1, α7-nAChR and parvalbumin, were prevented by NAC. CONCLUSION Our results highlights the influences of sex in NAC preventive effects in rats exposed to a two-hit schizophrenia model.
Collapse
Affiliation(s)
- Aline Santos Monte
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Francisco Eliclécio Rodrigues da Silva
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Camila Nayane de Carvalho Lima
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Germana Silva Vasconcelos
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Nayana Soares Gomes
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Fábio Miyajima
- Fundação Oswaldo Cruz (Fiocruz-CE), Fortaleza, Ceara, Brazil
| | - Silvania Maria Mendes Vasconcelos
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Clarissa S Gama
- Laboratory of Molecular Psychiatry, Hospital de Clínicas de Porto Alegre, Programa de Pós-Graduação em Psiquiatria e Ciências do Comportamento, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Mary V Seeman
- Department of Psychiatry, University of Toronto, ON, Canada
| | - David Freitas de Lucena
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Danielle S Macedo
- Drug Research and Development Center, Department of Physiology and Pharmacology, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
- National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, SP, Brazil
| |
Collapse
|
42
|
Schroeder A, Nakamura JP, Hudson M, Jones NC, Du X, Sundram S, Hill RA. Raloxifene recovers effects of prenatal immune activation on cognitive task-induced gamma power. Psychoneuroendocrinology 2019; 110:104448. [PMID: 31546114 DOI: 10.1016/j.psyneuen.2019.104448] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/04/2019] [Accepted: 09/13/2019] [Indexed: 12/13/2022]
Abstract
There is currently no treatment available for the cognitive symptoms of schizophrenia, but evidence suggests that selective estrogen receptor modulators (SERMs) may provide relief. Our recent animal model data showed that a lack of female sex hormones in mice impairs the ability of hippocampal neurons to synchronise and generate oscillations within the frequency range of 30-80 Hz (gamma power) leading to cognitive impairment, while both estradiol and the SERM, raloxifene, recovered this. Given that cognitive impairment is accompanied by abnormal gamma power in schizophrenia, this study aimed to determine the effects of raloxifene on gamma power during spatial memory tasks in the prenatal immune challenged (poly-I:C) mouse model with relevance to schizophrenia. Pregnant dams received the viral mimetic poly-I:C (20 mg/kg, i.p.) at gestational day 17. Male and female offspring were treated with placebo or raloxifene implants at adulthood. Local field potentials from the CA1 hippocampus were simultaneously recorded during the Y-maze test of short term spatial memory and the cheeseboard maze test of long-term spatial learning and memory and cognitive flexibility. In female but not male mice, poly I:C exposure reduced gamma power during decision making and prolonged the time spent in the centre (decision making phase) during the Y-maze task. Female poly-I:C exposed mice also showed increased gamma power during acquisition of the cheeseboard long term memory task and perseverative behaviour. Treatment with raloxifene recovered gamma power and decision making deficits in the Y-maze and restored gamma power changes during the cheeseboard maze task as well as perseverative behaviour. Male mice showed no electrophysiological or behavioural effects of poly-I:C or raloxifene treatment. In summary, poly-I:C exposure induced female specific cognitive impairments accompanied by altered neural oscillations in the gamma frequency and raloxifene recovered these abnormalities.
Collapse
Affiliation(s)
- Anna Schroeder
- Psychoneuroendocrinology Laboratory, Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3010, Australia; Behavioural Neuroscience Laboratory, Department of Psychiatry, School of Clinical Sciences, Monash University, 246 Clayton Rd, Clayton, VIC, 3168, Australia
| | - Jay P Nakamura
- Psychoneuroendocrinology Laboratory, Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3010, Australia; Behavioural Neuroscience Laboratory, Department of Psychiatry, School of Clinical Sciences, Monash University, 246 Clayton Rd, Clayton, VIC, 3168, Australia
| | - Matthew Hudson
- Department of Medicine, University of Melbourne, Parkville, VIC, 3010, Australia; Department of Neuroscience, Monash University, Clayton, VIC, 3168, Australia
| | - Nigel C Jones
- Department of Medicine, University of Melbourne, Parkville, VIC, 3010, Australia; Department of Neuroscience, Monash University, Clayton, VIC, 3168, Australia
| | - Xin Du
- Psychoneuroendocrinology Laboratory, Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3010, Australia; Behavioural Neuroscience Laboratory, Department of Psychiatry, School of Clinical Sciences, Monash University, 246 Clayton Rd, Clayton, VIC, 3168, Australia
| | - Suresh Sundram
- Behavioural Neuroscience Laboratory, Department of Psychiatry, School of Clinical Sciences, Monash University, 246 Clayton Rd, Clayton, VIC, 3168, Australia
| | - Rachel A Hill
- Psychoneuroendocrinology Laboratory, Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 3010, Australia; Behavioural Neuroscience Laboratory, Department of Psychiatry, School of Clinical Sciences, Monash University, 246 Clayton Rd, Clayton, VIC, 3168, Australia.
| |
Collapse
|
43
|
Balit T, Abdel-Wahhab MA, Radenahmad N. Young Coconut Juice Reduces Some Histopathological Changes Associated with Alzheimer's Disease through the Modulation of Estrogen Receptors in Orchidectomized Rat Brains. J Aging Res 2019; 2019:7416419. [PMID: 31885921 PMCID: PMC6914913 DOI: 10.1155/2019/7416419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/06/2019] [Accepted: 09/16/2019] [Indexed: 11/23/2022] Open
Abstract
Propose. This study aimed to evaluate the protective role of young coconut juice (YCJ) against the pathological changes in Alzheimer's disease (AD) in orchidectomized (orx) rats. Methods and Results. Animals were divided into 7 groups including: baseline normal control group, sham control, orx rat group, orx rat group injected with 2.5 μg/kg b.w. estradiol benzoate (EB) 3 days a week for 10 weeks, and the orx rat groups treated orally with 10, 20, and 40 ml/kg b.w. of YCJ for 10 weeks. At the end of treatment period, animals were sacrificed and the brain of each rat was removed, fixed in 10% neutral formalin, and stained by specific antibodies against NF200, parvalbumin (PV), β-amyloid (Aβ), and estrogen receptors (ERα and ERβ). The results showed that the number of NF200- and PV-reactive neurons in the hippocampus and cerebral cortex was significantly reduced in orx rats. However, it restored to normal in orx rats injected with EB or those administrated with YCJ in a dose-related manner. Neurons containing β-amyloid (Aβ), a hallmark of Alzheimer's disease (AD), were found to be increased in the orx rats; however; they were reduced by EB injection or YCJ administration. These results suggested the binding of the YCJ active ingredient(s) with estrogen receptors (ERs) in the brain as indicated by the detection of ERα and ERβ in neurons since a significant correlation was detected between NF200-/PV-reactive neurons vs ERα-/ERβ-reactive neurons.Conclusion. It could be concluded that YCJ is effective as EB in reducing AD pathology, probably by being selective estrogen receptor modulators.
Collapse
Affiliation(s)
- Tatcha Balit
- Department of Anatomy, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Mosaad A. Abdel-Wahhab
- Department of Food Toxicology and Contaminants, National Research Center, Dokki, Cairo, Egypt
| | - Nisaudah Radenahmad
- Department of Anatomy, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| |
Collapse
|
44
|
Klinger K, Gomes FV, Rincón-Cortés M, Grace AA. Female rats are resistant to the long-lasting neurobehavioral changes induced by adolescent stress exposure. Eur Neuropsychopharmacol 2019; 29:1127-1137. [PMID: 31371105 PMCID: PMC6773464 DOI: 10.1016/j.euroneuro.2019.07.134] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/30/2019] [Accepted: 07/15/2019] [Indexed: 01/10/2023]
Abstract
Stress during adolescence is a risk factor for neuropsychiatric diseases, including schizophrenia. We recently observed that peripubertal male rats exposed to a combination of daily footshock plus restraint stress exhibited schizophrenia-like changes. However, numerous studies have shown sex differences in neuropsychiatric diseases as well as on the impact of coping with stress. Thus, we decided to evaluate, in adolescent female rats, the impact of different stressors (restraint stress [RS], footshock [FS], or the combination of FS and RS [FS+RS]) on social interaction (3-chamber social approach test/SAT), anxiety responses (elevated plus-maze/EPM), cognitive function (novel object recognition test/NOR), and dopamine (DA) system responsivity by evaluating locomotor response to amphetamine and in vivo extracellular single unit recordings of DA neurons in the ventral tegmental area (VTA) in adulthood. The impact of FS+RS during early adulthood was also investigated. Adolescent stress had no impact on social behavior, anxiety, cognition and locomotor response to amphetamine. In addition, adolescent stress did not induce long-lasting changes in VTA DA system activity. However, a decrease in the firing rate of VTA DA neurons was found 1-2 weeks post-adolescent stress. Similar to adolescent stress, adult stress did not induce long-lasting behavioral deficits and changes in VTA DA system activity, but FS+RS decreased VTA DA neuron population activity 1-2 weeks post-adult stress. Our results are consistent with previous studies showing that female rodents appear to be more resilient to developmental stress-induced persistent changes than males and may contribute to the delayed onset and lesser severity of schizophrenia in females.
Collapse
Affiliation(s)
- Katharina Klinger
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA; Institute of Genetic and Molecular Neurobiology, Otto-von-Guericke University, University of Magdeburg, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Felipe V Gomes
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA; Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, 3900 Bandeirantes Ave, Ribeirao Preto, SP, 14049-900, Brazil
| | - Millie Rincón-Cortés
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA.
| |
Collapse
|
45
|
Page CE, Coutellier L. Prefrontal excitatory/inhibitory balance in stress and emotional disorders: Evidence for over-inhibition. Neurosci Biobehav Rev 2019; 105:39-51. [PMID: 31377218 DOI: 10.1016/j.neubiorev.2019.07.024] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 01/04/2023]
Abstract
Chronic stress-induced emotional disorders like anxiety and depression involve imbalances between the excitatory glutamatergic system and the inhibitory GABAergic system in the prefrontal cortex (PFC). However, the precise nature and trajectory of excitatory/inhibitory (E/I) imbalances in these conditions is not clear, with the literature reporting glutamatergic and GABAergic findings that are at times contradictory and inconclusive. Here we propose and discuss the hypothesis that chronic stress-induced emotional dysfunction involves hypoactivity of the PFC due to increased inhibition. We will also discuss E/I imbalances in the context of sex differences. In this review, we will synthesize research about how glutamatergic and GABAergic systems are perturbed by chronic stress and in related emotional disorders like anxiety and depression and propose ideas for reconciling contradictory findings in support of the hypothesis of over-inhibition. We will also discuss evidence for how aspects of the GABAergic system such as parvalbumin (PV) cells can be targeted therapeutically for reinstating activity and plasticity in the PFC and treating stress-related disorders.
Collapse
Affiliation(s)
- Chloe E Page
- Department of Neuroscience, Ohio State University, Columbus OH, United States
| | - Laurence Coutellier
- Department of Neuroscience, Ohio State University, Columbus OH, United States; Department of Psychology, Ohio State University, Columbus OH, United States.
| |
Collapse
|
46
|
Heermann T, Garrett L, Wurst W, Fuchs H, Gailus-Durner V, Hrabě de Angelis M, Graw J, Hölter SM. Crybb2 Mutations Consistently Affect Schizophrenia Endophenotypes in Mice. Mol Neurobiol 2019; 56:4215-4230. [PMID: 30291584 DOI: 10.1007/s12035-018-1365-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/25/2018] [Indexed: 10/28/2022]
Abstract
As part of the βγ-superfamily, βB2-crystallin (CRYBB2) is an ocular structural protein in the lens, and mutation of the corresponding gene can cause cataracts. CRYBB2 also is expressed in non-lens tissue such as the adult mouse brain and is associated with neuropsychiatric disorders such as schizophrenia. Nevertheless, the robustness of this association as well as how CRYBB2 may contribute to disease-relevant phenotypes is unknown. To add further clarity to this issue, we performed a comprehensive analysis of behavioral and neurohistological alterations in mice with an allelic series of mutations in the C-terminal end of the Crybb2 gene. Behavioral phenotyping of these three βB2-mutant lines Crybb2O377, Crybb2Philly, and Crybb2Aey2 included assessment of exploratory activity and anxiety-related behavior in the open field, sensorimotor gating measured by prepulse inhibition (PPI) of the acoustic startle reflex, cognitive performance measured by social discrimination, and spontaneous alternation in the Y-maze. In each mutant line, we also quantified the number of parvalbumin-positive (PV+) GABAergic interneurons in selected brain regions that express CRYBB2. While there were allele-specific differences in individual behaviors and affected brain areas, all three mutant lines exhibited consistent alterations in PPI that paralleled alterations in the PV+ cell number in the thalamic reticular nucleus (TRN). The direction of the PPI change mirrored that of the TRN PV+ cell number thereby suggesting a role for TRN PV+ cell number in modulating PPI. Moreover, as both altered PPI and PV+ cell number are schizophrenia-associated endophenotypes, our result implicates mutated Crybb2 in the development of this neuropsychiatric disorder.
Collapse
Affiliation(s)
- Tamara Heermann
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany
- Max Planck Institute of Biochemistry, Munich, Germany
| | - Lillian Garrett
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany
- Developmental Genetics, Technische Universität München- Weihenstephan, c/o Helmholtz Zentrum München, Munich, Germany
- German Centre of Neurodegenerative Diseases (DZNE), Site Munich, Feodor-Lynen-Str. 17, 81377, Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Adolf-Butenandt-Institut, Ludwig-Maximilians-Universität München, Schillerstr.44, 80336, Munich, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany
| | - Valerie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany
- Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, 85354, Freising, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Jochen Graw
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany
| | - Sabine M Hölter
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany.
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Centre for Environmental Health, 85764, Neuherberg, Germany.
| |
Collapse
|
47
|
Du X, McCarthny CR, Notaras M, van den Buuse M, Hill RA. Effect of adolescent androgen manipulation on psychosis-like behaviour in adulthood in BDNF heterozygous and control mice. Horm Behav 2019; 112:32-41. [PMID: 30928609 DOI: 10.1016/j.yhbeh.2019.03.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 02/27/2019] [Accepted: 03/25/2019] [Indexed: 11/27/2022]
Abstract
RATIONALE Males are more prone to psychosis, schizophrenia and substance abuse and addiction in adolescence and early adulthood than females. However, the role of androgens during this developmental period is poorly understood. OBJECTIVES This study aimed to examine how androgens in adolescence influence psychosis-like behaviour in adulthood and whether brain-derived neurotrophic factor (BDNF) is a mediator of these developmental effects. METHODS Wild-type and BDNF heterozygous male mice were castrated at pre-pubescence and implanted with testosterone or dihydrotestosterone (DHT). In adulthood, we assessed amphetamine- and MK-801-induced hyperlocomotion as a model of psychosis-like behaviour. Western blot analysis was used to quantify levels of the dopamine transporter (DAT) and N-methyl-d-aspartate (NMDA) receptor subunits. RESULTS While castration itself had little effect on behaviour, adolescent testosterone, but not DHT, significantly reduced amphetamine-induced hyperlocomotion, whereas both testosterone and DHT reduced the effect of MK-801. These effects were similar in mice of either genotype. In wildtype mice, both testosterone and DHT treatment reduced DAT expression in the medial prefrontal cortex (mPFC) but these effects were absent in BDNF heterozygous mice. There were no effects on NMDA receptor subunit levels. CONCLUSIONS The differential effect of adolescent testosterone and DHT on amphetamine-induced hyperlocomotion in adulthood suggests involvement of conversion of testosterone to estrogen and subsequent modulation of dopaminergic signalling. In contrast, the similar effect of testosterone and DHT treatment on NMDA receptor-mediated hyperlocomotion indicates it is mediated by androgen receptors. The involvement of BDNF in these hormone effects remains to be elucidated. These results demonstrate that, during adolescence, androgens significantly influence key pathways related to various mental illnesses prevalent in adolescence.
Collapse
Affiliation(s)
- X Du
- Department of Psychiatry, Monash University, Melbourne, Australia
| | - C R McCarthny
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Australia
| | - M Notaras
- Center for Neurogenetics, Brain & Mind Research Institute, Cornell University, NY, USA
| | - M van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia; Department of Pharmacology, University of Melbourne, Australia; The College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia.
| | - R A Hill
- Department of Psychiatry, Monash University, Melbourne, Australia
| |
Collapse
|
48
|
Griffiths BB, Madden AMK, Edwards KA, Zup SL, Stary CM. Age-dependent sexual dimorphism in hippocampal cornu ammonis-1 perineuronal net expression in rats. Brain Behav 2019; 9:e01265. [PMID: 30912298 PMCID: PMC6520292 DOI: 10.1002/brb3.1265] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 12/21/2018] [Accepted: 02/12/2019] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Perineuronal nets (PNNs) are extracellular matrices that encompass parvalbumin-expressing parvalbumin positive (PVALB+) fast-spiking inhibitory interneurons where they protect and stabilize afferent synapses. Recent observations that gonadal hormones influence PVALB+ neuron development suggest that PNN regulation may be sexually dimorphic. Sex differences in PNN abundance and complexity have been reported in sexually dimorphic nuclei in zebra finch brains; however, corresponding differences in mammalian brains have not been investigated. METHODS In this study we assessed the number of cortical and hippocampal PNNs in juvenile and young adult male and female rats using fluorescent immunohistochemistry for PVALB and the PNN marker Wisteria Floribunda Lectin. RESULTS We report here that PNNs are numerous and well developed in hippocampal cornu ammonis-1 of adult males but are lower in juvenile and possibly adult females. No significant differences were observed between sexes in cornu ammonis-3 or adjacent neocortex. There was an observed developmental difference in the neocortex as juveniles had more PVALB+ cells, but fewer PNN+ cells, than adults. CONCLUSIONS Because PNNs are integral for several hippocampal-mediated learning and memory tasks, these observations have potential sex-dependent translational implications for clinical strategies targeting cognitive dysfunction.
Collapse
Affiliation(s)
- Brian B Griffiths
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, California
| | - Amanda M K Madden
- Developmental and Brain Sciences Program, University of Massachusetts Boston, Boston, Massachusetts
| | - Kimbra A Edwards
- Developmental and Brain Sciences Program, University of Massachusetts Boston, Boston, Massachusetts.,Department of Psychology, University of Massachusetts Boston, Boston, Massachusetts
| | - Susan L Zup
- Developmental and Brain Sciences Program, University of Massachusetts Boston, Boston, Massachusetts.,Department of Psychology, University of Massachusetts Boston, Boston, Massachusetts
| | - Creed M Stary
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, California
| |
Collapse
|
49
|
Du X, Hill RA. Hypothalamic-pituitary-gonadal axis dysfunction: An innate pathophysiology of schizophrenia? Gen Comp Endocrinol 2019; 275:38-43. [PMID: 30753842 DOI: 10.1016/j.ygcen.2019.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 02/08/2019] [Accepted: 02/08/2019] [Indexed: 12/21/2022]
Abstract
The female hormone 17β-estradiol is postulated to be protective against schizophrenia onset and severity. Hypoestrogenism is a common phenomenon in women with schizophrenia that has serious effects that adds to the burden of an already very onerous disease. The cause of hypoestrogenism is largely attributed to antipsychotic-induced hyperprolactinemia. Evidence suggest however that a significant portion of female schizophrenia patients develop hypoestrogenism either before antipsychotic treatment or without regard to the level of prolactin, suggesting that for a sizeable segment of female patients, gonadal abnormality may be an innate and early aspect of the disease. This review aims to summarise the available literature that examines gonadal dysfunction in schizophrenia through this prism as well as to outline some recent developments in treatment strategies that may provide feasible ways to successfully tackle hypoestrogenism in schizophrenia.
Collapse
Affiliation(s)
- X Du
- Behavioural Neuroscience Laboratory, Department of Psychiatry, Monash University, Clayton, Victoria 3168, Australia.
| | - R A Hill
- Behavioural Neuroscience Laboratory, Department of Psychiatry, Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
50
|
Thériault RK, Perreault ML. Hormonal regulation of circuit function: sex, systems and depression. Biol Sex Differ 2019; 10:12. [PMID: 30819248 PMCID: PMC6394099 DOI: 10.1186/s13293-019-0226-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 02/18/2019] [Indexed: 01/10/2023] Open
Abstract
Major depressive disorder (MDD) is a debilitating chronic illness that is two times more prevalent in women than in men. The mechanisms associated with the increased female susceptibility to depression remain poorly characterized. Aberrant neuronal oscillatory activity within the putative depression network is an emerging mechanism underlying MDD. However, innate sex differences in network activity and its contribution to depression vulnerability have not been well described. In this review, current evidence of sex differences in neuronal oscillatory activity, including the influence of sex hormones and female cycling, will first be described followed by evidence of disrupted neuronal circuit function in MDD and the effects of antidepressant treatment. Lastly, current knowledge of sex differences in MDD-associated aberrant circuit function and oscillatory activity will be highlighted, with an emphasis on the role of sex steroids and female cycling. Collectively, it is clear that there are significant gaps in the literature regarding innate and pathologically associated sex differences in network activity and that the elucidation of these differences is invaluable to our understanding of sex-specific vulnerabilities and therapies for MDD.
Collapse
Affiliation(s)
- Rachel-Karson Thériault
- Department of Molecular and Cellular Biology, University of Guelph (ON), 50 Stone Rd. E, Guelph, Ontario N1G 2W1 Canada
- Collaborative Neuroscience Program, University of Guelph (ON), Guelph, Canada
| | - Melissa L. Perreault
- Department of Molecular and Cellular Biology, University of Guelph (ON), 50 Stone Rd. E, Guelph, Ontario N1G 2W1 Canada
- Collaborative Neuroscience Program, University of Guelph (ON), Guelph, Canada
| |
Collapse
|