1
|
Martins LA, Schiavo A, Paz LV, Xavier LL, Mestriner RG. Neural underpinnings of fine motor skills under stress and anxiety: A review. Physiol Behav 2024; 282:114593. [PMID: 38782244 DOI: 10.1016/j.physbeh.2024.114593] [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: 03/05/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 05/25/2024]
Abstract
This review offers a comprehensive examination of how stress and anxiety affect motor behavior, particularly focusing on fine motor skills and gait adaptability. We explore the role of several neurochemicals, including brain-derived neurotrophic factor (BDNF) and dopamine, in modulating neural plasticity and motor control under these affective states. The review highlights the importance of developing therapeutic strategies that enhance motor performance by leveraging the interactions between key neurochemicals. Additionally, we investigate the complex interplay between emotional-cognitive states and sensorimotor behaviors, showing how stress and anxiety disrupt neural integration, leading to impairments in skilled movements and negatively impacting quality of life. Synthesizing evidence from human and rodent studies, we provide a detailed understanding of the relationships among stress, anxiety, and motor behavior. Our findings reveal neurophysiological pathways, behavioral outcomes, and potential therapeutic targets, emphasizing the intricate connections between neurobiological mechanisms, environmental factors, and motor performance.
Collapse
Affiliation(s)
- Lucas Athaydes Martins
- Pontifical Catholic University of Rio Grande do Sul (PUCRS). Graduate Program in Biomedical Gerontology, Av. Ipiranga, 6681, Porto Alegre, Brazil; Pontifical Catholic University of Rio Grande do Sul (PUCRS). Neuroscience, Motor Behavior, and Rehabilitation Research Group (NECORE-CNPq), Av. Ipiranga, 6681, Porto Alegre, Brazil
| | - Aniuska Schiavo
- Pontifical Catholic University of Rio Grande do Sul (PUCRS). Graduate Program in Biomedical Gerontology, Av. Ipiranga, 6681, Porto Alegre, Brazil; Pontifical Catholic University of Rio Grande do Sul (PUCRS). Neuroscience, Motor Behavior, and Rehabilitation Research Group (NECORE-CNPq), Av. Ipiranga, 6681, Porto Alegre, Brazil
| | - Lisiê Valéria Paz
- Pontifical Catholic University of Rio Grande do Sul (PUCRS). Graduate Program in Cellular and Molecular Biology, Av. Ipiranga, 6681, Porto Alegre, Brazil
| | - Léder Leal Xavier
- Pontifical Catholic University of Rio Grande do Sul (PUCRS). Neuroscience, Motor Behavior, and Rehabilitation Research Group (NECORE-CNPq), Av. Ipiranga, 6681, Porto Alegre, Brazil; Pontifical Catholic University of Rio Grande do Sul (PUCRS). Graduate Program in Cellular and Molecular Biology, Av. Ipiranga, 6681, Porto Alegre, Brazil
| | - Régis Gemerasca Mestriner
- Pontifical Catholic University of Rio Grande do Sul (PUCRS). Graduate Program in Biomedical Gerontology, Av. Ipiranga, 6681, Porto Alegre, Brazil; Pontifical Catholic University of Rio Grande do Sul (PUCRS). Neuroscience, Motor Behavior, and Rehabilitation Research Group (NECORE-CNPq), Av. Ipiranga, 6681, Porto Alegre, Brazil; Pontifical Catholic University of Rio Grande do Sul (PUCRS). Graduate Program in Cellular and Molecular Biology, Av. Ipiranga, 6681, Porto Alegre, Brazil.
| |
Collapse
|
2
|
Tsimpolis A, Kalafatakis K, Charalampopoulos I. Recent advances in the crosstalk between the brain-derived neurotrophic factor and glucocorticoids. Front Endocrinol (Lausanne) 2024; 15:1362573. [PMID: 38645426 PMCID: PMC11027069 DOI: 10.3389/fendo.2024.1362573] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
Brain-derived neurotrophic factor (BDNF), a key neurotrophin within the brain, by selectively activating the TrkB receptor, exerts multimodal effects on neurodevelopment, synaptic plasticity, cellular integrity and neural network dynamics. In parallel, glucocorticoids (GCs), vital steroid hormones, which are secreted by adrenal glands and rapidly diffused across the mammalian body (including the brain), activate two different groups of intracellular receptors, the mineralocorticoid and the glucocorticoid receptors, modulating a wide range of genomic, epigenomic and postgenomic events, also expressed in the neural tissue and implicated in neurodevelopment, synaptic plasticity, cellular homeostasis, cognitive and emotional processing. Recent research evidences indicate that these two major regulatory systems interact at various levels: they share common intracellular downstream pathways, GCs differentially regulate BDNF expression, under certain conditions BDNF antagonises the GC-induced effects on long-term potentiation, neuritic outgrowth and cellular death, while GCs regulate the intraneuronal transportation and the lysosomal degradation of BDNF. Currently, the BDNF-GC crosstalk features have been mainly studied in neurons, although initial findings show that this crosstalk could be equally important for other brain cell types, such as astrocytes. Elucidating the precise neurobiological significance of BDNF-GC interactions in a tempospatial manner, is crucial for understanding the subtleties of brain function and dysfunction, with implications for neurodegenerative and neuroinflammatory diseases, mood disorders and cognitive enhancement strategies.
Collapse
Affiliation(s)
- Alexandros Tsimpolis
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas (IMBB-FORTH), Heraklion, Greece
| | - Konstantinos Kalafatakis
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Faculty of Medicine and Dentistry (Malta Campus), Queen Mary University of London, Victoria, Malta
| | - Ioannis Charalampopoulos
- Department of Pharmacology, Medical School, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas (IMBB-FORTH), Heraklion, Greece
| |
Collapse
|
3
|
Vinnakota C, Schroeder A, Du X, Ikeda K, Ide S, Mishina M, Hudson M, Jones NC, Sundram S, Hill RA. Understanding the role of the NMDA receptor subunit, GluN2D, in mediating NMDA receptor antagonist-induced behavioral disruptions in male and female mice. J Neurosci Res 2024; 102:e25257. [PMID: 37814998 PMCID: PMC10953441 DOI: 10.1002/jnr.25257] [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: 05/08/2023] [Revised: 07/24/2023] [Accepted: 09/23/2023] [Indexed: 10/11/2023]
Abstract
Noncompetitive NMDA receptor (NMDAR) antagonists like phencyclidine (PCP) and ketamine cause psychosis-like symptoms in healthy humans, exacerbate schizophrenia symptoms in people with the disorder, and disrupt a range of schizophrenia-relevant behaviors in rodents, including hyperlocomotion. This is negated in mice lacking the GluN2D subunit of the NMDAR, suggesting the GluN2D subunit mediates the hyperlocomotor effects of these drugs. However, the role of GluN2D in mediating other schizophrenia-relevant NMDAR antagonist-induced behavioral disturbances, and in both sexes, is unclear. This study aimed to investigate the role of the GluN2D subunit in mediating schizophrenia-relevant behaviors induced by a range of NMDA receptor antagonists. Using both male and female GluN2D knockout (KO) mice, we examined the effects of the NMDAR antagonist's PCP, the S-ketamine enantiomer (S-ket), and the ketamine metabolite R-norketamine (R-norket) on locomotor activity, anxiety-related behavior, and recognition and short-term spatial memory. GluN2D-KO mice showed a blunted locomotor response to R-norket, S-ket, and PCP, a phenotype present in both sexes. GluN2D-KO mice of both sexes showed an anxious phenotype and S-ket, R-norket, and PCP showed anxiolytic effects that were dependent on sex and genotype. S-ket disrupted spatial recognition memory in females and novel object recognition memory in both sexes, independent of genotype. This datum identifies a role for the GluN2D subunit in sex-specific effects of NMDAR antagonists and on the differential effects of the R- and S-ket enantiomers.
Collapse
Affiliation(s)
- Chitra Vinnakota
- Department of PsychiatryMonash UniversityClaytonVictoriaAustralia
| | - Anna Schroeder
- Department of PsychiatryMonash UniversityClaytonVictoriaAustralia
| | - Xin Du
- Department of PsychiatryMonash UniversityClaytonVictoriaAustralia
| | - Kazutaka Ikeda
- Addictive Substance ProjectTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Soichiro Ide
- Addictive Substance ProjectTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Masayoshi Mishina
- Brain Science Laboratory, The Research Organization of Science and TechnologyRitsumeikan UniversityKusatsuJapan
| | - Matthew Hudson
- Department of NeuroscienceMonash UniversityClaytonVictoriaAustralia
| | | | - Suresh Sundram
- Department of PsychiatryMonash UniversityClaytonVictoriaAustralia
- Mental Health ProgramMonash HealthClaytonVictoriaAustralia
| | - Rachel Anne Hill
- Department of PsychiatryMonash UniversityClaytonVictoriaAustralia
| |
Collapse
|
4
|
Hogarth S, Jaehne EJ, Xu X, Schwarz Q, van den Buuse M. Interaction of Brain-Derived Neurotrophic Factor with the Effects of Chronic Methamphetamine on Prepulse Inhibition in Mice Is Independent of Dopamine D3 Receptors. Biomedicines 2023; 11:2290. [PMID: 37626786 PMCID: PMC10452514 DOI: 10.3390/biomedicines11082290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
The aim of the present study was to gain a better understanding of the role of brain-derived neurotrophic factor (BDNF) and dopamine D3 receptors in the effects of chronic methamphetamine (METH) on prepulse inhibition (PPI), an endophenotype of psychosis. We compared the effect of a three-week adolescent METH treatment protocol on the regulation of PPI in wildtype mice, BDNF heterozygous mice (HET), D3 receptor knockout mice (D3KO), and double-mutant mice (DM) with both BDNF heterozygosity and D3 receptor knockout. Chronic METH induced disruption of PPI regulation in male mice with BDNF haploinsufficiency (HET and DM), independent of D3 receptor knockout. Specifically, these mice showed reduced baseline PPI, as well as attenuated disruption of PPI induced by acute treatment with the dopamine receptor agonist, apomorphine (APO), or the glutamate NMDA receptor antagonist, MK-801. In contrast, there were no effects of BDNF heterozygosity or D3 knockout on PPI regulation in female mice. Chronic METH pretreatment induced the expected locomotor hyperactivity sensitisation, where female HET and DM mice also showed endogenous sensitisation. Differential sex-specific effects of genotype and METH pretreatment were observed on dopamine receptor and dopamine transporter gene expression in the striatum and frontal cortex. Taken together, these results show a significant involvement of BDNF in the long-term effects of METH on PPI, particularly in male mice, but these effects appear independent of D3 receptors. The role of this receptor in psychosis endophenotypes therefore remains unclear.
Collapse
Affiliation(s)
- Samuel Hogarth
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC 3086, Australia (E.J.J.)
| | - Emily J. Jaehne
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC 3086, Australia (E.J.J.)
| | - Xiangjun Xu
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5000, Australia (Q.S.)
| | - Quenten Schwarz
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5000, Australia (Q.S.)
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC 3086, Australia (E.J.J.)
| |
Collapse
|
5
|
Schonfeld L, Jaehne EJ, Ogden AR, Spiers JG, Hogarth S, van den Buuse M. Differential effects of chronic adolescent glucocorticoid or methamphetamine on drug-induced locomotor hyperactivity and disruption of prepulse inhibition in adulthood in mice. Prog Neuropsychopharmacol Biol Psychiatry 2022; 117:110552. [PMID: 35337859 DOI: 10.1016/j.pnpbp.2022.110552] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/17/2022] [Accepted: 03/17/2022] [Indexed: 11/29/2022]
Abstract
Sensitization of dopaminergic activity has been suggested as an underlying mechanism in the psychotic symptoms of schizophrenia. Adolescent stress and chronic abuse of methamphetamine (Meth) are well-known risk factors for psychosis and schizophrenia; however it remains unknown how these factors compare in terms of dopaminergic behavioural sensitization in adulthood. In addition, while Brain-Derived Neurotrophic Factor (BDNF) has been implicated in dopaminergic activity and schizophrenia, its role in behavioural sensitization remains unclear. In this study we therefore compared the effect of chronic adolescent treatment with the stress hormone, corticosterone (Cort), or with Meth, on drug-induced locomotor hyperactivity and disruption of prepulse inhibition in adulthood in BDNF heterozygous mice and their wild-type controls, as well as on dopamine receptor gene expression. Between 6 and 9 weeks of age, the animals either received Cort in the drinking water or were treated with an escalating Meth dose protocol. In adulthood, Cort-pretreated mice showed significantly reduced Meth-induced locomotor hyperactivity compared to vehicle-pretreated mice. In contrast, Meth hyperlocomotion was significantly enhanced in animals pretreated with the drug in adolescence. There were no effects of either pretreatment on prepulse inhibition. BDNF Het mice showed greater Meth-induced hyperlocomotion and lower prepulse inhibition than WT mice. There were no effects of either pretreatment on D1 or D2 gene expression in either the dorsal or ventral striatum, while D3 mRNA was shown to be reduced in male mice only irrespective of genotype. These results suggest that in adolescence, chronically elevated glucocorticoid levels, a component of chronic stress, do not cause dopaminergic sensitization adulthood, in contrast to the effect of chronic Meth treatment in the same age period. BDNF does not appear to be involved in the effects of chronic Cort or chronic Meth.
Collapse
Affiliation(s)
- Lina Schonfeld
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| | - Emily J Jaehne
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| | - Alexandra R Ogden
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| | - Jereme G Spiers
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Australia
| | - Samuel Hogarth
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| | - Maarten van den Buuse
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Melbourne, Australia; Department of Pharmacology, University of Melbourne, Australia; College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia.
| |
Collapse
|
6
|
Caradonna SG, Zhang TY, O’Toole N, Shen MJ, Khalil H, Einhorn NR, Wen X, Parent C, Lee FS, Akil H, Meaney MJ, McEwen BS, Marrocco J. Genomic modules and intramodular network concordance in susceptible and resilient male mice across models of stress. Neuropsychopharmacology 2022; 47:987-999. [PMID: 34848858 PMCID: PMC8938529 DOI: 10.1038/s41386-021-01219-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/28/2021] [Accepted: 10/18/2021] [Indexed: 12/24/2022]
Abstract
The multifactorial etiology of stress-related disorders necessitates a constant interrogation of the molecular convergences in preclinical models of stress that use disparate paradigms as stressors spanning from environmental challenges to genetic predisposition to hormonal signaling. Using RNA-sequencing, we investigated the genomic signatures in the ventral hippocampus common to mouse models of stress. Chronic oral corticosterone (CORT) induced increased anxiety- and depression-like behavior in wild-type male mice and male mice heterozygous for the gene coding for brain-derived neurotrophic factor Val66Met, a variant associated with genetic susceptibility to stress. In a separate set of male mice, chronic social defeat stress (CSDS) led to a susceptible or a resilient population, whose proportion was dependent on housing conditions, namely standard housing or enriched environment. Rank-rank-hypergeometric overlap (RRHO), a threshold-free approach that ranks genes by their p value and effect size direction, was used to identify genes from a continuous gradient of significancy that were concordant across groups. In mice treated with CORT and in standard-housed susceptible mice, differentially expressed genes (DEGs) were concordant for gene networks involved in neurotransmission, cytoskeleton function, and vascularization. Weighted gene co-expression analysis generated 54 gene hub modules and revealed two modules in which both CORT and CSDS-induced enrichment in DEGs, whose function was concordant with the RRHO predictions, and correlated with behavioral resilience or susceptibility. These data showed transcriptional concordance across models in which the stress coping depends upon hormonal, environmental, or genetic factors revealing common genomic drivers that embody the multifaceted nature of stress-related disorders.
Collapse
Affiliation(s)
- Salvatore G. Caradonna
- grid.134907.80000 0001 2166 1519Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY USA
| | - Tie-Yuan Zhang
- grid.14709.3b0000 0004 1936 8649Douglas Mental Health University Institute, McGill University, Montreal, QC Canada
| | - Nicholas O’Toole
- grid.14709.3b0000 0004 1936 8649Douglas Mental Health University Institute, McGill University, Montreal, QC Canada
| | - Mo-Jun Shen
- grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences, Singapore, Singapore
| | - Huzefa Khalil
- grid.214458.e0000000086837370Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI USA
| | - Nathan R. Einhorn
- grid.134907.80000 0001 2166 1519Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY USA
| | - Xianglan Wen
- grid.14709.3b0000 0004 1936 8649Douglas Mental Health University Institute, McGill University, Montreal, QC Canada
| | - Carine Parent
- grid.14709.3b0000 0004 1936 8649Douglas Mental Health University Institute, McGill University, Montreal, QC Canada
| | - Francis S. Lee
- grid.5386.8000000041936877XDepartment of Psychiatry, Sackler Institute for Developmental Psychobiology, Weill Cornell Medical College, New York, NY USA
| | - Huda Akil
- grid.214458.e0000000086837370Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI USA
| | - Michael J. Meaney
- grid.14709.3b0000 0004 1936 8649Douglas Mental Health University Institute, McGill University, Montreal, QC Canada ,grid.452264.30000 0004 0530 269XSingapore Institute for Clinical Sciences, Singapore, Singapore ,grid.4280.e0000 0001 2180 6431Yong Loo Lin School of Medicine, Singapore, Singapore ,grid.14709.3b0000 0004 1936 8649Sackler Program for Epigenetics & Psychobiology, McGill University, Montreal, QC Canada
| | - Bruce S. McEwen
- grid.134907.80000 0001 2166 1519Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY USA
| | - Jordan Marrocco
- Laboratory of Neuroendocrinology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
7
|
Corticosterone induces discrete epigenetic signatures in the dorsal and ventral hippocampus that depend upon sex and genotype: focus on methylated Nr3c1 gene. Transl Psychiatry 2022; 12:109. [PMID: 35296634 PMCID: PMC8927334 DOI: 10.1038/s41398-022-01864-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/17/2022] [Accepted: 02/24/2022] [Indexed: 02/07/2023] Open
Abstract
The genomic effects of circulating glucocorticoids are particularly relevant in cortico-limbic structures, which express a high concentration of steroid hormone receptors. To date, no studies have investigated genomic differences in hippocampal subregions, namely the dorsal (dHPC) and ventral (vHPC) hippocampus, in preclinical models treated with exogenous glucocorticoids. Chronic oral corticosterone (CORT) in mouse is a pharmacological approach that disrupts the activity of the hypothalamic-pituitary-adrenal axis, increases affective behavior, and induces genomic changes after stress in the HPC of wildtype (WT) mice and mice heterozygous for the gene coding for brain-derived neurotrophic factor Val66Met (hMet), a variant associated with genetic susceptibility to stress. Using RNA-sequencing, we investigated the genomic signatures of oral CORT in the dHPC and vHPC of WT and hMet male and female mice, and examined sex and genotype differences in response to oral CORT. Males under CORT showed lower glycemia and increased anxiety- and depression-like behavior compared to females that showed instead opposite affective behavior in response to CORT. Rank-rank-hypergeometric overlap (RRHO) was used to identify genes from a continuous gradient of significancy that were concordant across groups. RRHO showed that CORT-induced differentially expressed genes (DEGs) in WT mice and hMet mice converged in the dHPC of males and females, while in the vHPC, DEGs converged in males and diverged in females. The vHPC showed a higher number of DEGs compared to the dHPC and exhibited sex differences related to glucocorticoid receptor (GR)-binding genes and epigenetic modifiers. Methyl-DNA-immunoprecipitation in the vHPC revealed differential methylation of the exons 1C and 1F of the GR gene (Nr3c1) in hMet females. Together, we report behavioral and endocrinological sex differences in response to CORT, as well as epigenetic signatures that i) differ in the dHPC and vHPC,ii) are distinct in males and females, and iii) implicate differential methylation of Nr3c1 selectively in hMet females.
Collapse
|
8
|
Microbiota and Pain: Save Your Gut Feeling. Cells 2022; 11:cells11060971. [PMID: 35326422 PMCID: PMC8946251 DOI: 10.3390/cells11060971] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/28/2022] [Accepted: 03/08/2022] [Indexed: 12/04/2022] Open
Abstract
Recently, a growing body of evidence has emerged regarding the interplay between microbiota and the nervous system. This relationship has been associated with several pathological conditions and also with the onset and regulation of pain. Dysregulation of the axis leads to a huge variety of diseases such as visceral hypersensitivity, stress-induced hyperalgesia, allodynia, inflammatory pain and functional disorders. In pain management, probiotics have shown promising results. This narrative review describes the peripheral and central mechanisms underlying pain processing and regulation, highlighting the role of the gut-brain axis in the modulation of pain. We summarized the main findings in regard to the stress impact on microbiota’s composition and its influence on pain perception. We also focused on the relationship between gut microbiota and both visceral and inflammatory pain and we provided a summary of the main evidence regarding the mechanistic effects and probiotics use.
Collapse
|
9
|
Abstract
Most psychiatric illnesses, such as schizophrenia, show profound sex differences in incidence, clinical presentation, course, and outcome. Fortunately, more recently the literature on sex differences and (to a lesser extent) effects of sex steroid hormones is expanding, and in this review we have focused on such studies in psychosis, both from a clinical/epidemiological and preclinical/animal model perspective. We begin by briefly describing the clinical evidence for sex differences in schizophrenia epidemiology, symptomatology, and pathophysiology. We then detail sex differences and sex hormone effects in behavioral animal models of psychosis, specifically psychotropic drug-induced locomotor hyperactivity and disruption of prepulse inhibition. We expand on the preclinical data to include developmental and genetic models of psychosis, such as the maternal immune activation model and neuregulin transgenic animals, respectively. Finally, we suggest several recommendations for future studies, in order to facilitate a better understanding of sex differences in the development of psychosis.
Collapse
|
10
|
Guerrin CGJ, Doorduin J, Sommer IE, de Vries EFJ. The dual hit hypothesis of schizophrenia: Evidence from animal models. Neurosci Biobehav Rev 2021; 131:1150-1168. [PMID: 34715148 DOI: 10.1016/j.neubiorev.2021.10.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 10/21/2021] [Accepted: 10/24/2021] [Indexed: 12/16/2022]
Abstract
Schizophrenia is a heterogeneous psychiatric disorder, which can severely impact social and professional functioning. Epidemiological and clinical studies show that schizophrenia has a multifactorial aetiology comprising genetic and environmental risk factors. Although several risk factors have been identified, it is still not clear how they result in schizophrenia. This knowledge gap, however, can be investigated in animal studies. In this review, we summarise animal studies regarding molecular and cellular mechanisms through which genetic and environmental factors may affect brain development, ultimately causing schizophrenia. Preclinical studies suggest that early environmental risk factors can affect the immune, GABAergic, glutamatergic, or dopaminergic system and thus increase the susceptibility to another risk factor later in life. A second insult, like social isolation, stress, or drug abuse, can further disrupt these systems and the interactions between them, leading to behavioural abnormalities. Surprisingly, first insults like maternal infection and early maternal separation can also have protective effects. Single gene mutations associated with schizophrenia did not have a major impact on the susceptibility to subsequent environmental hits.
Collapse
Affiliation(s)
- Cyprien G J Guerrin
- Department of Nuclear Medicine and Medical Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Janine Doorduin
- Department of Nuclear Medicine and Medical Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Iris E Sommer
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands
| | - Erik F J de Vries
- Department of Nuclear Medicine and Medical Imaging, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713, GZ, Groningen, the Netherlands.
| |
Collapse
|
11
|
Sun W, Cheng H, Yang Y, Tang D, Li X, An L. Requirements of Postnatal proBDNF in the Hippocampus for Spatial Memory Consolidation and Neural Function. Front Cell Dev Biol 2021; 9:678182. [PMID: 34336832 PMCID: PMC8319730 DOI: 10.3389/fcell.2021.678182] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/03/2021] [Indexed: 11/17/2022] Open
Abstract
Mature brain-derived neurotrophic factor (BDNF) and its downstream signaling pathways have been implicated in regulating postnatal development and functioning of rodent brain. However, the biological role of its precursor pro-brain-derived neurotrophic factor (proBDNF) in the postnatal brain remains unknown. The expression of hippocampal proBDNF was blocked in postnatal weeks, and multiple behavioral tests, Western blot and morphological techniques, and neural recordings were employed to investigate how proBDNF played a role in spatial cognition in adults. The peak expression and its crucial effects were found in the fourth but not in the second or eighth postnatal week. Blocking proBDNF expression disrupted spatial memory consolidation rather than learning or memory retrieval. Structurally, blocking proBDNF led to the reduction in spine density and proportion of mature spines. Although blocking proBDNF did not affect N-methyl-D-aspartate (NMDA) receptor (NMDAR) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subunits, the learning-induced phosphorylation of the GluN2B subunit level declined significantly. Functionally, paired-pulse facilitation, post-low-frequency stimulation (LFS) transiently enhanced depression, and GluN2B-dependent short-lasting long-term depression in the Schaffer collateral-CA1 pathway were weakened. The firing rate of pyramidal neurons was significantly suppressed around the target region during the memory test. Furthermore, the activation of GluN2B-mediated signaling could effectively facilitate neural function and mitigate memory impairment. The findings were consistent with the hypothesis that postnatal proBDNF played an essential role in synaptic and cognitive functions.
Collapse
Affiliation(s)
- Wei Sun
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Hong Cheng
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Neurology, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yang Yang
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Dongxin Tang
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xiaolian Li
- Department of Neurology, Jinan Geriatric Hospital, Jinan, China
| | - Lei An
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Neurology, Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Physiology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
12
|
Harb M, Jagusch J, Durairaja A, Endres T, Leßmann V, Fendt M. BDNF haploinsufficiency induces behavioral endophenotypes of schizophrenia in male mice that are rescued by enriched environment. Transl Psychiatry 2021; 11:233. [PMID: 33888685 PMCID: PMC8062437 DOI: 10.1038/s41398-021-01365-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/26/2021] [Accepted: 04/12/2021] [Indexed: 02/02/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is implicated in a number of processes that are crucial for healthy functioning of the brain. Schizophrenia is associated with low BDNF levels in the brain and blood, however, not much is known about BDNF's role in the different symptoms of schizophrenia. Here, we used BDNF-haploinsufficient (BDNF+/-) mice to investigate the role of BDNF in different mouse behavioral endophenotypes of schizophrenia. Furthermore, we assessed if an enriched environment can prevent the observed changes. In this study, male mature adult wild-type and BDNF+/- mice were tested in mouse paradigms for cognitive flexibility (attentional set shifting), sensorimotor gating (prepulse inhibition), and associative emotional learning (safety and fear conditioning). Before these tests, half of the mice had a 2-month exposure to an enriched environment, including running wheels. After the tests, BDNF brain levels were quantified. BDNF+/- mice had general deficits in the attentional set-shifting task, increased startle magnitudes, and prepulse inhibition deficits. Contextual fear learning was not affected but safety learning was absent. Enriched environment housing completely prevented the observed behavioral deficits in BDNF+/- mice. Notably, the behavioral performance of the mice was negatively correlated with BDNF protein levels. These novel findings strongly suggest that decreased BDNF levels are associated with several behavioral endophenotypes of schizophrenia. Furthermore, an enriched environment increases BDNF protein to wild-type levels and is thereby able to rescue these behavioral endophenotypes.
Collapse
Affiliation(s)
- Mahmoud Harb
- grid.5807.a0000 0001 1018 4307Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Justina Jagusch
- grid.5807.a0000 0001 1018 4307Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Archana Durairaja
- grid.5807.a0000 0001 1018 4307Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Thomas Endres
- grid.5807.a0000 0001 1018 4307Institute of Physiology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Volkmar Leßmann
- Institute of Physiology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany. .,Center for Behavioral Brain Sciences, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.
| | - Markus Fendt
- Institute for Pharmacology and Toxicology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany. .,Center for Behavioral Brain Sciences, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.
| |
Collapse
|
13
|
van den Buuse M, Halley P, Hoyne GF. Behavioural phenotyping of thunder mice with a hypomorphic mutation of heterogeneous nuclear ribonuclear protein L-like (hnRNPLL) and reduced T cell function. Neurosci Lett 2020; 740:135469. [PMID: 33152455 DOI: 10.1016/j.neulet.2020.135469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/23/2020] [Accepted: 10/27/2020] [Indexed: 02/01/2023]
Abstract
Heterogeneous nuclear ribonuclear protein l-like (hnRNPLL) is an RNA binding protein that regulates alternative splicing of mRNA and is abundantly expressed in memory T lymphocytes of the immune system and in the brain. A hypomorphic allele of the gene encoding hnRNPLL (Hnrpllthunder) selectively reduces T cell accumulation in lymphoid tissues, but little is known about its effects in the brain. Therefore, we exposed Hnrpllthunder mice to a test battery with relevance for a range of psychiatric illnesses. Thunder mice showed enhanced immobility in the tail-suspension test for depression-related behaviours, impaired short-term spatial memory in the Y-maze and reduced avoidance learning in the active avoidance test. Thus, in addition to its reported effects on immune function, the hnRNPLL mutation in thunder mice selectively affected aspects of behaviour.
Collapse
Affiliation(s)
- Maarten van den Buuse
- Mental Health Research Institute of Victoria, Parkville, VIC, Australia; School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia.
| | - Paul Halley
- Mental Health Research Institute of Victoria, Parkville, VIC, Australia
| | - Gerard F Hoyne
- Immunology Program, John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia; School of Health Sciences, University of Notre Dame Australia, Fremantle, WA, Australia
| |
Collapse
|
14
|
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: 183] [Impact Index Per Article: 45.8] [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
|
15
|
Bistoletti M, Bosi A, Banfi D, Giaroni C, Baj A. The microbiota-gut-brain axis: Focus on the fundamental communication pathways. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 176:43-110. [PMID: 33814115 DOI: 10.1016/bs.pmbts.2020.08.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Michela Bistoletti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Annalisa Bosi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Davide Banfi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, Varese, Italy.
| | - Andreina Baj
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| |
Collapse
|
16
|
Miao Z, Wang Y, Sun Z. The Relationships Between Stress, Mental Disorders, and Epigenetic Regulation of BDNF. Int J Mol Sci 2020; 21:ijms21041375. [PMID: 32085670 PMCID: PMC7073021 DOI: 10.3390/ijms21041375] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/01/2020] [Accepted: 02/15/2020] [Indexed: 12/25/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF), a critical member of the neurotrophic family, plays an important role in multiple stress-related mental disorders. Although alterations in BDNF in multiple brain regions of individuals experiencing stress have been demonstrated in previous studies, it appears that a set of elements are involved in the complex regulation. In this review, we summarize the specific brain regions with altered BDNF expression during stress exposure. How various environmental factors, including both physical and psychological stress, affect the expression of BDNF in specific brain regions are further summarized. Moreover, epigenetic regulation of BDNF, including DNA methylation, histone modification, and noncoding RNA, in response to diverse types of stress, as well as sex differences in the sensitivity of BDNF to the stress response, is also summarized. Clarification of the underlying role of BDNF in the stress process will promote our understanding of the pathology of stress-linked mental disorders and provide a potent target for the future treatment of stress-related illness.
Collapse
Affiliation(s)
- Zhuang Miao
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou 325000, China;
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China;
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yan Wang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China;
- School of Life Sciences, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Zhongsheng Sun
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou 325000, China;
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing 100101, China;
- School of Life Sciences, University of the Chinese Academy of Sciences, Beijing 100049, China
- Correspondence:
| |
Collapse
|
17
|
Notaras MJ, Vivian B, Wilson C, van den Buuse M. Interaction of reelin and stress on immobility in the forced swim test but not dopamine-mediated locomotor hyperactivity or prepulse inhibition disruption: Relevance to psychotic and mood disorders. Schizophr Res 2020; 215:485-492. [PMID: 28711473 DOI: 10.1016/j.schres.2017.07.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 07/03/2017] [Accepted: 07/06/2017] [Indexed: 12/28/2022]
Abstract
RATIONALE Psychotic disorders, such as schizophrenia, as well as some mood disorders, such as bipolar disorder, have been suggested to share common biological risk factors. One such factor is reelin, a large extracellular matrix glycoprotein that regulates neuronal migration during development as well as numerous activity-dependent processes in the adult brain. The current study sought to evaluate whether a history of stress exposure interacts with endogenous reelin levels to modify behavioural endophenotypes of relevance to psychotic and mood disorders. METHODS Heterozygous Reeler Mice (HRM) and wildtype (WT) controls were treated with 50mg/L of corticosterone (CORT) in their drinking water from 6 to 9weeks of age, before undergoing behavioural testing in adulthood. We assessed methamphetamine-induced locomotor hyperactivity, prepulse inhibition (PPI) of acoustic startle, short-term spatial memory in the Y-maze, and depression-like behaviour in the Forced-Swim Test (FST). RESULTS HRM genotype or CORT treatment did not affect methamphetamine-induced locomotor hyperactivity, a model of psychosis-like behaviour. At baseline, HRM showed decreased PPI at the commonly used 100msec interstimulus interval (ISI), but not at the 30msec ISI or following challenge with apomorphine. A history of CORT exposure potentiated immobility in the FST amongst HRM, but not WT mice. In the Y-maze, chronic CORT treatment decreased novel arm preference amongst HRM, reflecting reduced short-term spatial memory. CONCLUSION These data confirm a significant role of endogenous reelin levels on stress-related behaviour, supporting a possible role in both bipolar disorder and schizophrenia. However, an interaction of reelin deficiency with dopaminergic regulation of psychosis-like behaviour remains unclear.
Collapse
Affiliation(s)
- Michael J Notaras
- Florey Institute of Neuroscience & Mental Health, Melbourne, Victoria, Australia
| | - Billie Vivian
- School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Carey Wilson
- School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia; Department of Pharmacology, University of Melbourne, Victoria, Australia; The College of Public Health, Medical and Veterinary Sciences, James Cook University, Queensland, Australia.
| |
Collapse
|
18
|
Ieraci A, Herrera DG. Early Postnatal Ethanol Exposure in Mice Induces Sex-Dependent Memory Impairment and Reduction of Hippocampal NMDA-R2B Expression in Adulthood. Neuroscience 2019; 427:105-115. [PMID: 31874240 DOI: 10.1016/j.neuroscience.2019.11.045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/11/2022]
Abstract
Drinking alcohol during pregnancy is particularly detrimental for the developing brain and may cause a broad spectrum of cognitive and behavioral impairments, collectively known as fetal alcohol spectrum disorder (FASD). While behavioral abnormalities and brain damage have been widely investigated in animal models of FASD, the sex differences in the vulnerability to perinatal ethanol exposure have received less consideration. Here we investigated the long-term behavioral and molecular effects of acute ethanol-binge like exposure during the early postnatal period (equivalent to the third trimester of human pregnancy) in adult male and female mice. CD1 mice received a single ethanol exposure on P7 and were analyzed starting from P60. We found that ethanol-exposed mice showed increased activity in the open field test and in the plus-maze test, regardless of the sex. Interestingly, only ethanol-exposed adult male mice exhibited memory impairment in the water maze and fear-conditioning tests. Remarkably, hippocampal levels of NMDA-R2B were reduced only in ethanol-exposed male, while total BDNF levels were increased in both male and female ethanol-exposed mice. Our data suggest a different susceptibility of early postnatal ethanol exposure in male and female CD1 mice.
Collapse
Affiliation(s)
- Alessandro Ieraci
- Department of Psychiatry, Weill Medical College of Cornell University, New York, NY 10065, USA.
| | - Daniel G Herrera
- Department of Psychiatry, Weill Medical College of Cornell University, New York, NY 10065, USA
| |
Collapse
|
19
|
Sex-specific spatial memory deficits in mice with a conditional TrkB deletion on parvalbumin interneurons. Behav Brain Res 2019; 372:111984. [DOI: 10.1016/j.bbr.2019.111984] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/23/2019] [Accepted: 05/27/2019] [Indexed: 12/12/2022]
|
20
|
Involvement of brain-derived neurotrophic factor (BDNF) in the long-term memory effects of glucocorticoid stimulation during adolescence/young adulthood. Behav Brain Res 2019; 377:112223. [PMID: 31518662 DOI: 10.1016/j.bbr.2019.112223] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/09/2019] [Accepted: 09/09/2019] [Indexed: 12/22/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) has been implicated in cognition and the effects of chronic stress. We have previously shown in mice that chronic adolescent treatment with corticosterone (CORT), to simulate stress, resulted in spatial memory deficits and markedly elevated levels of the N-methyl-D-aspartate (NMDA) receptor subunit NR2B in adult male BDNF heterozygous mice (BDNF+/-), but not in wildtype controls (WT) or females. The aim of the present study was to further characterize this 'two hit' model, including whether these effects are long-lasting. CORT treatment was delivered in the drinking water from 6 to 9 weeks of age. As previously demonstrated, male BDNF+/- mice treated with CORT presented with a deficit in spatial memory at 11 weeks of age. However, this deficit was not maintained at 15 weeks of age. Conversely, male WT treated with CORT developed a deficit only at 15 weeks of age. There were no significant gene-environment interactions in female mice at any time point. CORT treatment caused a modest, but significant increase in NR2B levels which was independent of genotype. These results show marked age-dependent and sex-dependent effects of CORT on behaviour which are different in BDNF+/- mice than in controls.
Collapse
|
21
|
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: 0.8] [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
|
22
|
Glutamatergic Signaling Along The Microbiota-Gut-Brain Axis. Int J Mol Sci 2019; 20:ijms20061482. [PMID: 30934533 PMCID: PMC6471396 DOI: 10.3390/ijms20061482] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/04/2019] [Accepted: 03/21/2019] [Indexed: 12/21/2022] Open
Abstract
A complex bidirectional communication system exists between the gastrointestinal tract and the brain. Initially termed the “gut-brain axis” it is now renamed the “microbiota-gut-brain axis” considering the pivotal role of gut microbiota in maintaining local and systemic homeostasis. Different cellular and molecular pathways act along this axis and strong attention is paid to neuroactive molecules (neurotransmitters, i.e., noradrenaline, dopamine, serotonin, gamma aminobutyric acid and glutamate and metabolites, i.e., tryptophan metabolites), sustaining a possible interkingdom communication system between eukaryota and prokaryota. This review provides a description of the most up-to-date evidence on glutamate as a neurotransmitter/neuromodulator in this bidirectional communication axis. Modulation of glutamatergic receptor activity along the microbiota-gut-brain axis may influence gut (i.e., taste, visceral sensitivity and motility) and brain functions (stress response, mood and behavior) and alterations of glutamatergic transmission may participate to the pathogenesis of local and brain disorders. In this latter context, we will focus on two major gut disorders, such as irritable bowel syndrome and inflammatory bowel disease, both characterized by psychiatric co-morbidity. Research in this area opens the possibility to target glutamatergic neurotransmission, either pharmacologically or by the use of probiotics producing neuroactive molecules, as a therapeutic approach for the treatment of gastrointestinal and related psychiatric disorders.
Collapse
|
23
|
Li J, Li Y, Sun Y, Wang H, Liu X, Zhao Y, Wang H, Su Y, Si T. Chronic mild corticosterone exposure during adolescence enhances behaviors and upregulates neuroplasticity-related proteins in rat hippocampus. Prog Neuropsychopharmacol Biol Psychiatry 2019; 89:400-411. [PMID: 30392783 DOI: 10.1016/j.pnpbp.2018.10.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 10/24/2018] [Accepted: 10/26/2018] [Indexed: 11/16/2022]
Abstract
Adolescence is a critical period with ongoing maturational processes in stress-sensitive systems. It remains unknown how adolescent individuals would be affected by chronic exposure to corticosterone (the major stress hormone in rodents, CORT) at the doses that are usually not detrimental in adults. In this study, male Sprague-Dawley rats were injected with CORT (5 mg/kg) or vehicle for 21 days during adolescence (postnatal day (PND) 29-49) or adulthood (PND 71-91) and then subjected to behavioral testing or sacrifice for neurobiological analyses. Shortly after treatment cessation, different from CORT-treated adults showing increased anxiety-like behaviors, CORT-treated adolescents exhibited enhanced prepulse inhibition and spatial learning. They also showed increased expression of hippocampal neuroplasticity-related proteins, including BDNF, nectin3, and AMPA receptor subunits. These effects became undetectable after a four-week washout period when CORT-treated adolescents exhibited improved reversal learning. Together, these findings demonstrate that chronic CORT exposure at the dose of 5 mg/kg endows adolescent individuals with enhanced cognitive capacities, possibly supported by increased hippocampal neuroplasticity. This study also highlights mild elevation of CORT levels during adolescence as a potential approach of promoting adaptive behaviors.
Collapse
Affiliation(s)
- Jitao Li
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing 100191, China
| | - Youhong Li
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing 100191, China
| | - Yaxin Sun
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing 100191, China
| | - Han Wang
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing 100191, China
| | - Xiao Liu
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing 100191, China; The Sixth People's Hospital of Hebei Province, Baoding 071000, China
| | - Yingying Zhao
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing 100191, China; MECT Treatment Center, Beijing Anding Hospital of Capital Medical University, Beijing 100088, China
| | - Hongli Wang
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing 100191, China
| | - Yun'ai Su
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing 100191, China
| | - Tianmei Si
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University), Beijing 100191, China.
| |
Collapse
|
24
|
Notaras M, van den Buuse M. Brain-Derived Neurotrophic Factor (BDNF): Novel Insights into Regulation and Genetic Variation. Neuroscientist 2018; 25:434-454. [DOI: 10.1177/1073858418810142] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Since its discovery, brain-derived neurotrophic factor (BDNF) has spawned a literature that now spans 35 years of research. While all neurotrophins share considerable overlap in sequence homology and their processing, BDNF has become the most widely studied neurotrophin because of its broad roles in brain homeostasis, health, and disease. Although research on BDNF has produced thousands of articles, there remain numerous long-standing questions on aspects of BDNF molecular biology and signaling. Here we provide a comprehensive review, including both a historical narrative and a forward-looking perspective on advances in the actions of BDNF within the brain. We specifically review BDNF’s gene structure, peptide composition (including domains, posttranslational modifications and putative motif sites), mechanisms of transport, signaling pathway recruitment, and other recent developments including the functional effects of genetic variation and the discovery of a new BDNF prodomain ligand. This body of knowledge illustrates a highly conserved and complex role for BDNF within the brain, that promotes the idea that the neurotrophin biology of BDNF is diverse and that any disease involvement is likely to be equally multifarious.
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, Victoria, Australia
- College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Queensland, Australia
- Department of Pharmacology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
25
|
Grech AM, Ratnayake U, Hannan AJ, van den Buuse M, Hill RA. Sex-Dependent Effects of Environmental Enrichment on Spatial Memory and Brain-Derived Neurotrophic Factor (BDNF) Signaling in a Developmental "Two-Hit" Mouse Model Combining BDNF Haploinsufficiency and Chronic Glucocorticoid Stimulation. Front Behav Neurosci 2018; 12:227. [PMID: 30356704 PMCID: PMC6189322 DOI: 10.3389/fnbeh.2018.00227] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 09/11/2018] [Indexed: 01/02/2023] Open
Abstract
Neurodevelopmental disorders are thought to be caused by a combination of adverse genetic and environmental insults. The "two-hit" hypothesis suggests that an early first "hit" primes the developing brain to be vulnerable to a second "hit" during adolescence which triggers behavioral dysfunction. We have previously modeled this scenario in mice and found that the combined effect of a genetic hapolinsuffuciency in the brain-derived neurotrophic factor (BDNF) gene (1st hit) and chronic corticosterone (CORT) treatment during adolescence (2nd hit), caused spatial memory impairments in adulthood. Environmental enrichment (EE) protocols are designed to stimulate experience-dependent plasticity and have shown therapeutic actions. This study investigated whether EE can reverse these spatial memory impairments. Wild-type (WT) and BDNF heterozygous (HET) mice were treated with corticosterone (CORT) in their drinking water (50 mg/L) from weeks 6 to 8 and exposed to EE from 7 to 9 weeks. Enriched housing included open top cages with additional toys, tunnels, housing, and platforms. Y-maze novel preference testing, to assess short-term spatial memory, was performed at 10 weeks of age. At week 16 dorsal hippocampus tissue was obtained for Western blot analysis of expression levels of BDNF, the BDNF receptor TrkB, and NMDA receptor subunits, GluNR1, 2A and 2B. As in our previous studies, spatial memory was impaired in our two-hit (BDNF HET + CORT) mice. Simultaneous EE prevented these impairments. However, EE appeared to worsen spatial memory performance in WT mice, particularly those exposed to CORT. While BDNF levels were lower in BDNF HET mice as expected, there were no further effects of CORT or EE in males but a close to significant female CORT × EE × genotype interaction which qualitatively corresponded with Y-maze performance. However, EE caused both sex- and genotype-specific effects on phosphorylated TrkB residues and GluNR expression within the dorsal hippocampus, with GluNR2B levels in males changing in parallel with spatial memory performance. In conclusion, beneficial effects of EE on spatial memory emerge only following two developmental disruptions. The mechanisms by which EE exerts its effects are likely via regulation of multiple activity-dependent pathways, including TrkB and NMDA receptor signaling.
Collapse
Affiliation(s)
- Adrienne M. Grech
- Department of Psychiatry, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Udani Ratnayake
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Anthony J. Hannan
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
- Department of Pharmacology, University of Melbourne, Melbourne, VIC, Australia
- The College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
| | - Rachel A. Hill
- Department of Psychiatry, School of Clinical Sciences, Monash Medical Centre, Monash University, Clayton, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
26
|
Interaction of Brain-Derived Neurotrophic Factor Val66Met genotype and history of stress in regulation of prepulse inhibition in mice. Schizophr Res 2018; 198:60-67. [PMID: 28864281 DOI: 10.1016/j.schres.2017.08.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 08/11/2017] [Accepted: 08/15/2017] [Indexed: 12/22/2022]
Abstract
The Brain-Derived Neurotrophic Factor (BDNF) Val66Met polymorphism results in reduced activity-dependent BDNF release and has been implicated in schizophrenia. However, effects of the polymorphism on functional dopaminergic and N-methyl-d-aspartate (NMDA) receptor-associated activity remain unclear. We used prepulse inhibition, a measure of sensorimotor gating which is disrupted in schizophrenia, and assessed the effects of acute treatment with the dopamine receptor agonist, apomorphine (APO), and the NMDA receptor antagonist, MK-801. We used adult humanized hBDNFVal66Met 'knockin' mice which express either the Val/Val, Val/Met or Met/Met genotype. An interaction of BDNF with stress was modelled by chronic young-adult treatment with corticosterone (CORT). At 1 or 3mg/kg, APO had no effect in Val/Val mice but significantly reduced PPI at the 100ms inter-stimulus interval (ISI) in Val/Met and Met/Met mice. However, after CORT pretreatment, APO significantly reduced PPI in all genotypes similarly. At 0.1 or 0.25mg/kg, MK-801 significantly disrupted PPI at the 100ms ISI independent of genotype or CORT pretreatment. There were differential effects of APO and MK-801 on PPI at the 30ms ISI and startle between the genotypes, irrespective of CORT pretreatment. These results show that the BDNF Val66Met Val/Met and Met/Met genotypes are more sensitive than the Val/Val genotype to the effect of APO on PPI. A history of stress, here modelled by chronic CORT administration, increases effects of APO in Val/Val mice.
Collapse
|
27
|
On the Developmental Timing of Stress: Delineating Sex-Specific Effects of Stress across Development on Adult Behavior. Brain Sci 2018; 8:brainsci8070121. [PMID: 29966252 PMCID: PMC6071226 DOI: 10.3390/brainsci8070121] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/26/2018] [Accepted: 06/27/2018] [Indexed: 12/11/2022] Open
Abstract
Stress, and the chronic overactivation of major stress hormones, is associated with several neuropsychiatric disorders. However, clinical literature on the exact role of stress either as a causative, triggering, or modulatory factor to mental illness remains unclear. We suggest that the impact of stress on the brain and behavior is heavily dependent on the developmental timing at which the stress has occurred, and as such, this may contribute to the overall variability reported on the association of stress and mental illness. Here, animal models provide a way to comprehensively assess the temporal impact of stress on behavior in a controlled manner. This review particularly focuses on the long-term impact of stress on behavior in various rodent stress models at three major developmental time points: early life, adolescence, and adulthood. We characterize the various stressor paradigms into physical, social, and pharmacological, and discuss commonalities and differences observed across these various stress-inducing methods. In addition, we discuss here how sex can influence the impact of stress at various developmental time points. We conclude here that early postnatal life and adolescence represent particular periods of vulnerability, but that stress exposure during early life can sometimes lead to resilience, particularly to fear-potentiated memories. In the adult brain, while shorter periods of stress tended to enhance spatial memory, longer periods caused impairments. Overall, males tended to be more vulnerable to the long-term effects of early life and adolescent stress, albeit very few studies incorporate both sexes, and further well-powered sex comparisons are needed.
Collapse
|
28
|
Schroeder A, van den Buuse M, Hill RA. Reelin Haploinsufficiency and Late-Adolescent Corticosterone Treatment Induce Long-Lasting and Female-Specific Molecular Changes in the Dorsal Hippocampus. Brain Sci 2018; 8:brainsci8070118. [PMID: 29941797 PMCID: PMC6070826 DOI: 10.3390/brainsci8070118] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 06/20/2018] [Accepted: 06/22/2018] [Indexed: 12/24/2022] Open
Abstract
Reelin depletion and stress seem to affect similar pathways including GABAergic and glutamatergic signaling and both are implicated in psychiatric disorders in late adolescence/early adulthood. The interaction between reelin depletion and stress, however, remains unclear. To investigate this, male and female heterozygous reelin mice (HRM) and wildtype (WT) controls were treated with the stress hormone, corticosterone (CORT), during late adolescence to simulate chronic stress. Glucocorticoid receptors (GR), N-methyl-d-aspartate receptor (NMDAr) subunits, glutamic acid decarboxylase (GAD67) and parvalbumin (PV) were measured in the hippocampus and the prefrontal cortex (PFC) in adulthood. While no changes were seen in male mice, female HRM showed a significant reduction in GR expression in the dorsal hippocampus. In addition, CORT reduced GR levels as well as GluN2B and GluN2C subunits of NMDAr in the dorsal hippocampus in female mice only. CORT furthermore reduced GluN1 levels in the PFC of female mice. The combined effect of HRM and CORT treatment appeared to be additive in terms of GR expression in the dorsal hippocampus. Female-specific CORT-induced changes were associated with overall higher circulating CORT levels in female compared to male mice. This study shows differential effects of reelin depletion and CORT treatment on GR and NMDAr protein expression in male and female mice, suggesting that females are more susceptible to reelin haploinsufficiency as well as late-adolescent stress. These findings shed more light on female-specific vulnerability to stress and have implications for stress-associated mental illnesses with a female bias including anxiety and major depression.
Collapse
Affiliation(s)
- Anna Schroeder
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Australia.
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton 3168, Australia.
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Bundoora 3086 Australia.
- Department of Pharmacology, University of Melbourne, Parkville 3052, Australia.
- The College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville 4810, Australia.
| | - Rachel A Hill
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Australia.
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton 3168, Australia.
| |
Collapse
|
29
|
Du X, Serena K, Hwang WJ, Grech A, Wu Y, Schroeder A, Hill R. Prefrontal cortical parvalbumin and somatostatin expression and cell density increase during adolescence and are modified by BDNF and sex. Mol Cell Neurosci 2018; 88:177-188. [DOI: 10.1016/j.mcn.2018.02.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 01/25/2018] [Accepted: 02/01/2018] [Indexed: 01/21/2023] Open
|
30
|
Garner JM, Chambers J, Barnes AK, Datta S. Changes in Brain-Derived Neurotrophic Factor Expression Influence Sleep-Wake Activity and Homeostatic Regulation of Rapid Eye Movement Sleep. Sleep 2017; 41:4643005. [PMID: 29462410 PMCID: PMC6018753 DOI: 10.1093/sleep/zsx194] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Study Objectives Brain-derived neurotrophic factor (BDNF) expression and homeostatic regulation of rapid eye movement (REM) sleep are critical for neurogenesis and behavioral plasticity. Accumulating clinical and experimental evidence suggests that decreased BDNF expression is causally linked with the development of REM sleep-associated neuropsychiatric disorders. Therefore, we hypothesize that BDNF plays a role in sleep–wake (S–W) activity and homeostatic regulation of REM sleep. Methods Male and female wild-type (WT; BDNF +/+) and heterozygous BDNF (KD; BDNF +/−) rats were chronically implanted with S–W recording electrodes to quantify baseline S–W activity and REM sleep homeostatic regulatory processes during the light phase. Results Molecular analyses revealed that KD BDNF rats had a 50% decrease in BDNF protein levels. During baseline S–W activity, KD rats exhibited fewer REM sleep episodes that were shorter in duration and took longer to initiate. Also, the baseline S–W activity did not reveal any sex difference. During the 3-hour selective REM sleep deprivation, KD rats failed to exhibit a homeostatic drive for REM sleep and did not exhibit rebound REM sleep during the recovery S–W period. Conclusion Interestingly, both genotypes did not reveal any sex difference in the quality and/or quantity of REM sleep. Collectively, these results, for the first time, unequivocally demonstrate that an intact BDNF system in both sexes is a critical modulator for baseline and homeostatic regulation of REM sleep. This study further suggests that heterozygous BDNF knockdown rats are a useful animal model for the study of the cellular and molecular mechanisms of sleep regulation and cognitive functions of sleep.
Collapse
Affiliation(s)
- Jennifer M Garner
- Department of Anesthesiology, Graduate School of Medicine, University of Tennessee, Knoxville, TN.,Department of Psychology, College of Arts and Sciences, Knoxville, TN
| | - Jonathan Chambers
- Department of Anesthesiology, Graduate School of Medicine, University of Tennessee, Knoxville, TN
| | - Abigail K Barnes
- Department of Anesthesiology, Graduate School of Medicine, University of Tennessee, Knoxville, TN.,Department of Psychology, College of Arts and Sciences, Knoxville, TN
| | - Subimal Datta
- Department of Anesthesiology, Graduate School of Medicine, University of Tennessee, Knoxville, TN.,Department of Psychology, College of Arts and Sciences, Knoxville, TN.,Program in Comparative and Experimental Medicine; University of Tennessee, Knoxville, TN
| |
Collapse
|
31
|
Geist PA, Dulka BN, Barnes A, Totty M, Datta S. RETRACTED: BNDF heterozygosity is associated with memory deficits and alterations in cortical and hippocampal EEG power. Behav Brain Res 2017; 332:154-163. [PMID: 28576309 PMCID: PMC5534188 DOI: 10.1016/j.bbr.2017.05.039] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 05/02/2017] [Accepted: 05/11/2017] [Indexed: 12/31/2022]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Editor-in-Chief, the Corresponding Author and the Institutional Research Integrity office at the University of Tennessee due to data mis-management of the project, including inappropriate removal of animals from analyses and use of unapproved euthanasia.
Collapse
Affiliation(s)
- Phillip A Geist
- Department of Anesthesiology, Graduate School of Medicine, The University of Tennessee, 1924 Alcoa Highway, Knoxville, TN 37920, United States
| | - Brooke N Dulka
- Department of Psychology, College of Arts and Sciences, 1404 Circle Drive, United States
| | - Abigail Barnes
- Department of Anesthesiology, Graduate School of Medicine, The University of Tennessee, 1924 Alcoa Highway, Knoxville, TN 37920, United States; Department of Psychology, College of Arts and Sciences, 1404 Circle Drive, United States
| | - Michael Totty
- Department of Anesthesiology, Graduate School of Medicine, The University of Tennessee, 1924 Alcoa Highway, Knoxville, TN 37920, United States
| | - Subimal Datta
- Department of Anesthesiology, Graduate School of Medicine, The University of Tennessee, 1924 Alcoa Highway, Knoxville, TN 37920, United States; Department of Psychology, College of Arts and Sciences, 1404 Circle Drive, United States; Program in Comparative and Experimental Medicine, 2407 River Drive, RM A102, The University of Tennessee, Knoxville, TN 37996, United States.
| |
Collapse
|
32
|
Notaras MJ, Hill RA, Gogos JA, van den Buuse M. BDNF Val66Met Genotype Interacts With a History of Simulated Stress Exposure to Regulate Sensorimotor Gating and Startle Reactivity. Schizophr Bull 2017; 43:665-672. [PMID: 27262112 PMCID: PMC5464110 DOI: 10.1093/schbul/sbw077] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Reduced expression of Brain-Derived Neurotrophic Factor (BDNF) has been implicated in the pathophysiology of schizophrenia. The BDNF Val66Met polymorphism, which results in deficient activity-dependent secretion of BDNF, is associated with clinical features of schizophrenia. We investigated the effect of this polymorphism on Prepulse Inhibition (PPI), a translational model of sensorimotor gating which is disrupted in schizophrenia. We utilized humanized BDNFVal66Met (hBDNFVal66Met) mice which have been modified to carry the Val66Met polymorphism, as well as express humanized BDNF in vivo. We also studied the long-term effect of chronic corticosterone (CORT) exposure in these animals as a model of history of stress. PPI was assessed at 30ms and 100ms interstimulus intervals (ISI). Analysis of PPI at the commonly used 100ms ISI identified that, irrespective of CORT treatment, the hBDNFVal/Met genotype was associated with significantly reduced PPI. In contrast, PPI was not different between hBDNFMet/Met and hBDNFVal/Val genotype mice. At the 30ms ISI, CORT treatment selectively disrupted sensorimotor gating of hBDNFVal/Met heterozygote mice but not hBDNFVal/Val or hBDNFMet/Met mice. Analysis of startle reactivity revealed that chronic CORT reduced startle reactivity of hBDNFVal/Val male mice by 51%. However, this was independent of the effect of CORT on PPI. In summary, we provide evidence of a distinct BDNFVal66Met heterozygote-specific phenotype using the sensorimotor gating endophenotype of schizophrenia. These data have important implications for clinical studies where, if possible, the BDNFVal/Met heterozygote genotype should be distinguished from the BDNFMet/Met genotype.
Collapse
Affiliation(s)
- Michael J. Notaras
- Behavioural Neuroscience Laboratory, Florey Institute of Neuroscience and Mental Health, Melbourne, Australia;,Psychoneuroendocrinology Laboratory, Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| | - Rachel A. Hill
- Psychoneuroendocrinology Laboratory, Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| | - Joseph A. Gogos
- Departments of Biophysics and Neuroscience, Columbia University, New York, NY
| | - Maarten van den Buuse
- Behavioural Neuroscience Laboratory, Florey Institute of Neuroscience and Mental Health, Melbourne, Australia;,School of Psychology and Public Health, La Trobe University, Melbourne, Australia;,The College of Public Health, Medical and Veterinary Sciences, James Cook University, Queensland, Australia
| |
Collapse
|
33
|
van den Buuse M, Biel D, Radscheit K. Does genetic BDNF deficiency in rats interact with neurotransmitter control of prepulse inhibition? Implications for schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2017; 75:192-198. [PMID: 28192174 DOI: 10.1016/j.pnpbp.2017.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 02/06/2017] [Accepted: 02/08/2017] [Indexed: 01/19/2023]
Abstract
Several studies have suggested a role of BDNF in the development of schizophrenia. For example, post-mortem studies have shown significantly reduced levels of BDNF protein expression in the brain of schizophrenia patients. We investigated the relationship between reduced levels of BDNF in the brain and the regulation of prepulse inhibition (PPI), a behavioral endophenotype of schizophrenia. We used BDNF heterozygous mutant rats which display a 50% decrease of mature BDNF protein levels. Previously, we observed normal baseline PPI and responses to the dopamine D1/D2 receptor agonist, apomorphine, in these rats. Here, we focused on the effects of the NMDA receptor antagonist, MK-801, its interaction with mGluR2/3 and mGluR5 receptors, and the PPI response to serotonergic drugs. MK-801 administration caused a dose-dependent reduction of PPI and increase of startle amplitudes. Baseline PPI and the effect of 0.02-0.1mg/kg of MK-801 were not significantly altered in male or female BDNF heterozygous rats, although the MK-801-induced increase in startle levels was reduced. Co-treatment with the mGluR2/3 agonist, LY379,268, or the mGluR5 antagonist, MPEP, did not alter the effect of MK-801 on PPI in controls or BDNF mutant rats. Treatment with the serotonin-1A receptor agonist, 8-OH-DPAT, the serotonin-2A receptor agonist, DOI, or the serotonin releaser, fenfluramine, induced differential effects on PPI and startle but these effects were not different between the genotypes. These results show that a significant decrease of BDNF protein expression does not lead to reduced PPI at baseline or changes in the regulation of PPI via NMDA receptors or serotonergic mechanisms. These findings in a genetic rat model of BDNF deficiency do not support a role for similar reductions of BDNF levels in schizophrenia in the disruption of PPI, widely reported as an endophenotype of the illness. The potential implications of these results for our understanding of changes in PPI and BDNF expression in schizophrenia are discussed.
Collapse
Affiliation(s)
- Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia; Department of Pharmacology, University of Melbourne, Victoria, Australia; The College of Public Health, Medical and Veterinary Sciences, James Cook University, Queensland, Australia.
| | - Davina Biel
- School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia; Institute of Psychology, University of Luebeck, Luebeck, Germany
| | - Kathrin Radscheit
- School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
34
|
Li J, Xie X, Li Y, Liu X, Liao X, Su YA, Si T. Differential Behavioral and Neurobiological Effects of Chronic Corticosterone Treatment in Adolescent and Adult Rats. Front Mol Neurosci 2017; 10:25. [PMID: 28210212 PMCID: PMC5288376 DOI: 10.3389/fnmol.2017.00025] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 01/20/2017] [Indexed: 11/13/2022] Open
Abstract
Adolescence is a critical period with ongoing maturational processes in stress-sensitive systems. While adolescent individuals show heightened stress-induced hormonal responses compared to adults, it is unclear whether and how the behavioral and neurobiological consequences of chronic stress would differ between the two age groups. Here we address this issue by examining the effects of chronic exposure to the stress hormone, corticosterone (CORT), in both adolescent and adult animals. Male Sprague-Dawley (SD) rats were injected intraperitoneally with CORT (40 mg/kg) or vehicle for 21 days during adolescence (post-natal day (PND) 29–49) or adulthood (PND 71–91) and then subjected to behavioral testing or sacrifice for western blot analyses. Despite of similar physical and neuroendocrine effects in both age groups, chronic CORT treatment produced a series of behavioral and neurobiological effects with striking age differences. While CORT-treated adult animals exhibited decreased sucrose preference, increased anxiety levels and cognitive impairment, CORT-treated adolescent animals demonstrated increased sucrose preference, decreased anxiety levels, and increased sensorimotor gating functions. These differential behavioral alterations were accompanied by opposite changes in the two age groups in the expression levels of brain-derived neurotrophic factor (BDNF), the phosphorylation of the obligatory subunit of the NMDA receptor, GluN1, and PSD-95 in rat hippocampus. These results suggest that prolonged glucocorticoid exposure during adolescence produces different behavioral and neurobiological effects from those in adulthood, which may be due to the complex interaction between glucocorticoids and the ongoing neurodevelopmental processes during this period.
Collapse
Affiliation(s)
- Jitao Li
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University) Beijing, China
| | - Xiaomeng Xie
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University) Beijing, China
| | - Youhong Li
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University) Beijing, China
| | - Xiao Liu
- Department of Psychiatry and Mental Health, North China University of Science and Technology Tangshan, China
| | - Xuemei Liao
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University) Beijing, China
| | - Yun-Ai Su
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University) Beijing, China
| | - Tianmei Si
- National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital/Institute of Mental Health) and the Key Laboratory of Mental Health, Ministry of Health (Peking University) Beijing, China
| |
Collapse
|
35
|
A systematic review comparing sex differences in cognitive function in schizophrenia and in rodent models for schizophrenia, implications for improved therapeutic strategies. Neurosci Biobehav Rev 2016; 68:979-1000. [DOI: 10.1016/j.neubiorev.2016.06.029] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 06/11/2016] [Accepted: 06/20/2016] [Indexed: 01/07/2023]
|
36
|
The Gut-Brain Axis, BDNF, NMDA and CNS Disorders. Neurochem Res 2016; 41:2819-2835. [PMID: 27553784 DOI: 10.1007/s11064-016-2039-1] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/09/2016] [Accepted: 08/17/2016] [Indexed: 02/08/2023]
Abstract
Gastro-intestinal (GI) microbiota and the 'gut-brain axis' are proving to be increasingly relevant to early brain development and the emergence of psychiatric disorders. This review focuses on the influence of the GI tract on Brain-Derived Neurotrophic Factor (BDNF) and its relationship with receptors for N-methyl-D-aspartate (NMDAR), as these are believed to be involved in synaptic plasticity and cognitive function. NMDAR may be associated with the development of schizophrenia and a range of other psychopathologies including neurodegenerative disorders, depression and dementias. An analysis of the routes and mechanisms by which the GI microbiota contribute to the pathophysiology of BDNF-induced NMDAR dysfunction could yield new insights relevant to developing novel therapeutics for schizophrenia and related disorders. In the absence of GI microbes, central BDNF levels are reduced and this inhibits the maintenance of NMDAR production. A reduction of NMDAR input onto GABA inhibitory interneurons causes disinhibition of glutamatergic output which disrupts the central signal-to-noise ratio and leads to aberrant synaptic behaviour and cognitive deficits. Gut microbiota can modulate BDNF function in the CNS, via changes in neurotransmitter function by affecting modulatory mechanisms such as the kynurenine pathway, or by changes in the availability and actions of short chain fatty acids (SCFAs) in the brain. Interrupting these cycles by inducing changes in the gut microbiota using probiotics, prebiotics or antimicrobial drugs has been found promising as a preventative or therapeutic measure to counteract behavioural deficits and these may be useful to supplement the actions of drugs in the treatment of CNS disorders.
Collapse
|
37
|
Sex differences in animal models of schizophrenia shed light on the underlying pathophysiology. Neurosci Biobehav Rev 2016; 67:41-56. [DOI: 10.1016/j.neubiorev.2015.10.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/28/2015] [Accepted: 10/26/2015] [Indexed: 12/20/2022]
|
38
|
Elevated paternal glucocorticoid exposure alters the small noncoding RNA profile in sperm and modifies anxiety and depressive phenotypes in the offspring. Transl Psychiatry 2016; 6:e837. [PMID: 27300263 PMCID: PMC4931607 DOI: 10.1038/tp.2016.109] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 04/25/2016] [Indexed: 12/21/2022] Open
Abstract
Recent studies have suggested that physiological and behavioral traits may be transgenerationally inherited through the paternal lineage, possibly via non-genomic signals derived from the sperm. To investigate how paternal stress might influence offspring behavioral phenotypes, a model of hypothalamic-pituitary-adrenal (HPA) axis dysregulation was used. Male breeders were administered water supplemented with corticosterone (CORT) for 4 weeks before mating with untreated female mice. Female, but not male, F1 offspring of CORT-treated fathers displayed altered fear extinction at 2 weeks of age. Only male F1 offspring exhibited altered patterns of ultrasonic vocalization at postnatal day 3 and, as adults, showed decreased time in open on the elevated-plus maze and time in light on the light-dark apparatus, suggesting a hyperanxiety-like behavioral phenotype due to paternal CORT treatment. Interestingly, expression of the paternally imprinted gene Igf2 was increased in the hippocampus of F1 male offspring but downregulated in female offspring. Male and female F2 offspring displayed increased time spent in the open arm of the elevated-plus maze, suggesting lower levels of anxiety compared with control animals. Only male F2 offspring showed increased immobility time on the forced-swim test and increased latency to feed on the novelty-supressed feeding test, suggesting a depression-like phenotype in these animals. Collectively, these data provide evidence that paternal CORT treatment alters anxiety and depression-related behaviors across multiple generations. Analysis of the small RNA profile in sperm from CORT-treated males revealed marked effects on the expression of small noncoding RNAs. Sperm from CORT-treated males contained elevated levels of three microRNAs, miR-98, miR-144 and miR-190b, which are predicted to interact with multiple growth factors, including Igf2 and Bdnf. Sustained elevation of glucocorticoids is therefore involved in the transmission of paternal stress-induced traits across generations in a process involving small noncoding RNA signals transmitted by the male germline.
Collapse
|
39
|
Notaras M, Hill R, Gogos JA, van den Buuse M. BDNF Val66Met genotype determines hippocampus-dependent behavior via sensitivity to glucocorticoid signaling. Mol Psychiatry 2016; 21:730-2. [PMID: 26821977 PMCID: PMC4879187 DOI: 10.1038/mp.2015.152] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- M Notaras
- Behavioural Neuroscience Laboratory, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
- Psychoneuroendocrinology Laboratory, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - R Hill
- Psychoneuroendocrinology Laboratory, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
| | - J A Gogos
- Departments of Biophysics and Neuroscience, Columbia University, New York, NY, USA
| | - M van den Buuse
- Behavioural Neuroscience Laboratory, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC, Australia
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
40
|
Manning EE, van den Buuse M. Altered social cognition in male BDNF heterozygous mice and following chronic methamphetamine exposure. Behav Brain Res 2016; 305:181-5. [PMID: 26965573 DOI: 10.1016/j.bbr.2016.03.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 03/01/2016] [Accepted: 03/05/2016] [Indexed: 11/29/2022]
Abstract
Growing clinical evidence suggests that persistent psychosis which occurs in methamphetamine users is closely related to schizophrenia. However, preclinical studies in animal models have focussed on psychosis-related behaviours following methamphetamine, and less work has been done to assess endophenotypes relevant to other deficits observed in schizophrenia. Altered social behaviour is a feature of both the negative symptoms and cognitive deficits in schizophrenia, and significantly impacts patient functioning. We recently found that brain-derived neurotrophic factor (BDNF) heterozygous mice show disrupted sensitization to methamphetamine, supporting other work suggesting an important role of this neurotrophin in the pathophysiology of psychosis and the neuronal response to stimulant drugs. In the current study, we assessed social and cognitive behaviours in methamphetamine-treated BDNF heterozygous mice and wildtype littermate controls. Following chronic methamphetamine exposure male wildtype mice showed a 50% reduction in social novelty preference. Vehicle-treated male BDNF heterozygous mice showed a similar impairment in social novelty preference, with a trend for no further disruption by methamphetamine exposure. Female mice were unaffected in this task, and no groups showed any changes in sociability or short-term spatial memory. These findings suggest that chronic methamphetamine alters behaviour relevant to disruption of social cognition in schizophrenia, supporting other studies which demonstrate a close resemblance between persistent methamphetamine psychosis and schizophrenia. Together these findings suggest that dynamic regulation of BDNF signalling is necessary to mediate the effects of methamphetamine on behaviours relevant to schizophrenia.
Collapse
Affiliation(s)
- Elizabeth E Manning
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Australia; Translational OCD Laboratory, Department of Psychiatry, University of Pittsburgh, Pittsburgh, USA
| | - Maarten van den Buuse
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Australia; School of Psychology and Public Health, La Trobe University, Melbourne, Australia.
| |
Collapse
|
41
|
Manning EE, Halberstadt AL, van den Buuse M. BDNF-Deficient Mice Show Reduced Psychosis-Related Behaviors Following Chronic Methamphetamine. Int J Neuropsychopharmacol 2015; 19:pyv116. [PMID: 26453694 PMCID: PMC4851263 DOI: 10.1093/ijnp/pyv116] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/05/2015] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND One of the most devastating consequences of methamphetamine abuse is increased risk of psychosis. Brain-derived neurotrophic factor has been implicated in both psychosis and neuronal responses to methamphetamine. We therefore examined persistent psychosis-like behavioral effects of methamphetamine in brain-derived neurotrophic factor heterozygous mice. METHODS Mice were chronically treated with methamphetamine from 6 to 9 weeks of age, and locomotor hyperactivity to an acute D-amphetamine challenge was tested in photocell cages after a 2-week withdrawal period. RESULTS Methamphetamine-treated wild-type mice, but not brain-derived neurotrophic factor heterozygous mice, showed locomotor sensitization to acute 3mg/kg D-amphetamine. Qualitative analysis of exploration revealed tolerance to D-amphetamine effects on entropy in methamphetamine-treated brain-derived neurotrophic factor heterozygous mice, but not wild-type mice. CONCLUSIONS Chronic methamphetamine exposure induces contrasting profiles of behavioral changes in wild-type and brain-derived neurotrophic factor heterozygous mice, with attenuation of behaviors relevant to psychosis in methamphetamine-treated brain-derived neurotrophic factor heterozygous mice. This suggests that brain-derived neurotrophic factor signalling changes may contribute to development of psychosis in methamphetamine users.
Collapse
Affiliation(s)
| | | | - Maarten van den Buuse
- Florey Institute of Neuroscience and Mental Health Research, University of Melbourne, Victoria, Australia (Drs Manning and van den Buuse); Translational OCD Laboratory, Department of Psychiatry, University of Pittsburgh, PA (Dr Manning); Department of Psychiatry, University of California San Diego, La Jolla, CA (Dr Halberstadt); School of Psychology and Public Health, La Trobe University, Melbourne, Victoria, Australia (Dr van den Buuse).
| |
Collapse
|
42
|
Schroeder A, Buret L, Hill RA, van den Buuse M. Gene–environment interaction of reelin and stress in cognitive behaviours in mice: Implications for schizophrenia. Behav Brain Res 2015; 287:304-14. [DOI: 10.1016/j.bbr.2015.03.063] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 03/24/2015] [Accepted: 03/29/2015] [Indexed: 12/16/2022]
|
43
|
A role for the BDNF gene Val66Met polymorphism in schizophrenia? A comprehensive review. Neurosci Biobehav Rev 2015; 51:15-30. [DOI: 10.1016/j.neubiorev.2014.12.016] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 11/14/2014] [Accepted: 12/27/2014] [Indexed: 12/31/2022]
|
44
|
|
45
|
Petzold A, Psotta L, Brigadski T, Endres T, Lessmann V. Chronic BDNF deficiency leads to an age-dependent impairment in spatial learning. Neurobiol Learn Mem 2015; 120:52-60. [PMID: 25724412 DOI: 10.1016/j.nlm.2015.02.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 02/07/2015] [Accepted: 02/17/2015] [Indexed: 10/23/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a crucial mediator of neural plasticity and, consequently, of memory formation. In hippocampus-dependent learning tasks BDNF also seems to play an essential role. However, there are conflicting results concerning the spatial learning ability of aging BDNF(+/-) mice in the Morris water maze paradigm. To evaluate the effect of chronic BDNF deficiency in the hippocampus on spatial learning throughout life, we conducted a comprehensive study to test differently aged BDNF(+/-) mice and their wild type littermates in the Morris water maze and to subsequently quantify their hippocampal BDNF protein levels as well as expression levels of TrkB receptors. We observed an age-dependent learning deficit in BDNF(+/-) animals, starting at seven months of age, despite stable hippocampal BDNF protein expression and continual decline of TrkB receptor expression throughout aging. Furthermore, we detected a positive correlation between hippocampal BDNF protein levels and learning performance during the probe trial in animals that showed a good learning performance during the long-term memory test.
Collapse
Affiliation(s)
- Anne Petzold
- Institute for Physiology, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Laura Psotta
- Institute for Physiology, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Tanja Brigadski
- Institute for Physiology, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany
| | - Thomas Endres
- Institute for Physiology, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany.
| | - Volkmar Lessmann
- Institute for Physiology, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120 Magdeburg, Germany; Center for Behavioral Brain Sciences, Magdeburg, Germany
| |
Collapse
|
46
|
Samsom JN, Wong AHC. Schizophrenia and Depression Co-Morbidity: What We have Learned from Animal Models. Front Psychiatry 2015; 6:13. [PMID: 25762938 PMCID: PMC4332163 DOI: 10.3389/fpsyt.2015.00013] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 01/24/2015] [Indexed: 12/15/2022] Open
Abstract
Patients with schizophrenia are at an increased risk for the development of depression. Overlap in the symptoms and genetic risk factors between the two disorders suggests a common etiological mechanism may underlie the presentation of comorbid depression in schizophrenia. Understanding these shared mechanisms will be important in informing the development of new treatments. Rodent models are powerful tools for understanding gene function as it relates to behavior. Examining rodent models relevant to both schizophrenia and depression reveals a number of common mechanisms. Current models which demonstrate endophenotypes of both schizophrenia and depression are reviewed here, including models of CUB and SUSHI multiple domains 1, PDZ and LIM domain 5, glutamate Delta 1 receptor, diabetic db/db mice, neuropeptide Y, disrupted in schizophrenia 1, and its interacting partners, reelin, maternal immune activation, and social isolation. Neurotransmission, brain connectivity, the immune system, the environment, and metabolism emerge as potential common mechanisms linking these models and potentially explaining comorbid depression in schizophrenia.
Collapse
Affiliation(s)
- James N Samsom
- Department of Molecular Neuroscience, Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute , Toronto, ON , Canada ; Department of Pharmacology, Faculty of Medicine, University of Toronto , Toronto, ON , Canada
| | - Albert H C Wong
- Department of Molecular Neuroscience, Centre for Addiction and Mental Health, Campbell Family Mental Health Research Institute , Toronto, ON , Canada ; Department of Pharmacology, Faculty of Medicine, University of Toronto , Toronto, ON , Canada ; Department of Psychiatry, Faculty of Medicine, University of Toronto , Toronto, ON , Canada
| |
Collapse
|
47
|
Wu YWC, Du X, van den Buuse M, Hill RA. Analyzing the influence of BDNF heterozygosity on spatial memory response to 17β-estradiol. Transl Psychiatry 2015; 5:e498. [PMID: 25603414 PMCID: PMC4312832 DOI: 10.1038/tp.2014.143] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 12/02/2014] [Indexed: 12/25/2022] Open
Abstract
The recent use of estrogen-based therapies as adjunctive treatments for the cognitive impairments of schizophrenia has produced promising results; however the mechanism behind estrogen-based cognitive enhancement is relatively unknown. Brain-derived neurotrophic factor (BDNF) regulates learning and memory and its expression is highly responsive to estradiol. We recently found that estradiol modulates the expression of hippocampal parvalbumin-positive GABAergic interneurons, known to regulate neuronal synchrony and cognitive function. What is unknown is whether disruptions to the aforementioned estradiol-parvalbumin pathway alter learning and memory, and whether BDNF may mediate these events. Wild-type (WT) and BDNF heterozygous (+/-) mice were ovariectomized (OVX) at 5 weeks of age and simultaneously received empty, estradiol- or progesterone-filled implants for 7 weeks. At young adulthood, mice were tested for spatial and recognition memory in the Y-maze and novel-object recognition test, respectively. Hippocampal protein expression of BDNF and GABAergic interneuron markers, including parvalbumin, were assessed. WT OVX mice show impaired performance on Y-maze and novel-object recognition test. Estradiol replacement in OVX mice prevented the Y-maze impairment, a Behavioral abnormality of dorsal hippocampal origin. BDNF and parvalbumin protein expression in the dorsal hippocampus and parvalbumin-positive cell number in the dorsal CA1 were significantly reduced by OVX in WT mice, while E2 replacement prevented these deficits. In contrast, BDNF(+/-) mice showed either no response or an opposite response to hormone manipulation in both behavioral and molecular indices. Our data suggest that BDNF status is an important biomarker for predicting responsiveness to estrogenic compounds which have emerged as promising adjunctive therapeutics for schizophrenia patients.
Collapse
Affiliation(s)
- Y W C Wu
- Behavioural Neuroscience Laboratory, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, VIC, Australia
| | - X Du
- Behavioural Neuroscience Laboratory, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - M van den Buuse
- Behavioural Neuroscience Laboratory, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, VIC, Australia,School of Psychological Science, La Trobe University, Melbourne, VIC, Australia
| | - R A Hill
- Behavioural Neuroscience Laboratory, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia,Behavioural Neuroscience Laboratory, Florey Institute of Neuroscience and Mental Health, Kenneth Myer Building, Genetics Lane, Royal Parade, University of Melbourne, Parkville, VIC 3010, Australia. E-mail:
| |
Collapse
|
48
|
Brain-derived neurotrophic factor heterozygous mutant rats show selective cognitive changes and vulnerability to chronic corticosterone treatment. Neuroscience 2015; 284:297-310. [DOI: 10.1016/j.neuroscience.2014.10.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 09/11/2014] [Accepted: 10/08/2014] [Indexed: 01/08/2023]
|
49
|
Corticosterone treatment during adolescence induces down-regulation of reelin and NMDA receptor subunit GLUN2C expression only in male mice: implications for schizophrenia. Int J Neuropsychopharmacol 2014; 17:1221-32. [PMID: 24556017 DOI: 10.1017/s1461145714000121] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Stress exposure during adolescence/early adulthood has been shown to increase the risk for psychiatric disorders such as schizophrenia. Reelin plays an essential role in brain development and its levels are decreased in schizophrenia. However, the relationship between stress exposure and reelin expression remains unclear. We therefore treated adolescent reelin heteroyzogous mice (HRM) and wild-type (WT) littermates with the stress hormone, corticosterone (CORT) in their drinking water (25 mg/l) for 3 wk. In adulthood, we measured levels of full-length (FL) reelin and the N-R6 and N-R2 cleavage fragments in the frontal cortex (FC) and dorsal (DH) and ventral (VH) hippocampus. As expected, levels of all reelin forms were approximately 50% lower in HRMs compared to WT. In male mice, CORT treatment significantly decreased FL and N-R2 expression in the FC and N-R2 and N-R6 levels in the DH. This reelin down-regulation was accompanied by significant reductions in downstream N-methyl-D-aspartate (NMDA) GluN2C subunit levels. There were no effects of CORT treatment in the VH of either of the sexes and only subtle changes in female DH. CORT-induced reelin and GluN2C down-regulation in males was not associated with changes in two GABAergic neuron markers, GAD67 and parvalbumin, or glucocorticoids receptors (GR). These results show that CORT treatment causes long-lasting and selective reductions of reelin form levels in male FC and DH accompanied by changes in NMDAR subunit composition. This sex-specific reelin down-regulation in regions implicated in schizophrenia could be involved in the effects of stress in this disease.
Collapse
|
50
|
Gururajan A, Hill R, van den Buuse M. Long-term differential effects of chronic young-adult corticosterone exposure on anxiety and depression-like behaviour in BDNF heterozygous rats depend on the experimental paradigm used. Neurosci Lett 2014; 576:6-10. [DOI: 10.1016/j.neulet.2014.05.041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 05/16/2014] [Accepted: 05/19/2014] [Indexed: 12/13/2022]
|