101
|
Ai H, Shi XF, Hu XP, Fang WQ, Zhang B, Lu W. Acute stress regulates phosphorylation of N-methyl-d-aspartate receptor GluN2B at S1284 in hippocampus. Neuroscience 2017; 351:24-35. [PMID: 28359951 DOI: 10.1016/j.neuroscience.2017.03.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 03/07/2017] [Accepted: 03/19/2017] [Indexed: 01/07/2023]
Abstract
Exposure to acute stress leads to diverse changes, which include either beneficial or deleterious effects on molecular levels that are implicated in stress-related disorders. N-methyl-d-aspartate receptor (NMDAR)-mediated signalings, are thought to be vital players in stress-related mental disorders as well as attractive therapeutic targets for clinical treatment. In the present study, we utilized acute stress models in mice to explore regulation of phosphorylation level of S1284 in GluN2B subunit of NMDAR. We found out that forced swimming and acute restraint stress increased phosphorylation level of S1284, while phosphorylation level of S1284 was unaltered after brief exposure to open field. Moreover, phosphorylation change of S1284 was negated by treatment of roscovitine which is believed to be a Cyclin-dependent kinase inhibitor. Besides, we showed well correlation of phosphorylation change of S1284 and immobility time during forced swimming. Collectively, our results demonstrated that phosphorylation level of S1284 in GluN2B was regulated by acute stress.
Collapse
Affiliation(s)
- Heng Ai
- Department of Physiology, Hangzhou Medical College, Hangzhou, Zhejiang 310053, China
| | - Xiao-Fang Shi
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Xu-Pang Hu
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Wei-Qing Fang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Collaborative Innovation Center for Brain Science, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Bin Zhang
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Science, Hangzhou Normal University, Hangzhou, Zhejiang 310036, China
| | - Wen Lu
- Department of Biochemistry and Molecular Biology, Hainan Medical College, 571199, China.
| |
Collapse
|
102
|
Adrenocorticotropic hormone protects learning and memory function in epileptic Kcna1-null mice. Neurosci Lett 2017; 645:14-18. [PMID: 28249786 DOI: 10.1016/j.neulet.2017.02.069] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 02/10/2017] [Accepted: 02/25/2017] [Indexed: 11/20/2022]
Abstract
ACTH, a member of the melanocortin family of peptides, is often used in the treatment of the developmental epileptic encephalopathy spectrum disorders including, Ohtahara, West, Lennox Gastaut and Landau-Kleffner Syndromes and electrical status epilepticus of sleep. In these disorders, although ACTH is often successful in controlling the seizures and/or inter-ictal EEG abnormalities, it is unknown whether ACTH possesses other beneficial effects independent of seizure control. We tested whether ACTH can ameliorate the intrinsic impairment of hippocampal-based learning and memory in epileptic Kcna1-null (KO) mice. We found that ACTH - administered in the form of Acthar Gel given i.p. four times daily at a dose of 4 IU/kg (16 IU/kg/day) for 7days - prevented impairment of long-term potentiation (LTP) evoked with high-frequency stimulation in CA1 hippocampus and also restored spatial learning and memory on the Barnes maze test. However, with this treatment regimen, ACTH did not exert a significant effect on the frequency of spontaneous recurrent seizures. Together, our findings indicate that ACTH can ameliorate memory impairment in epileptic Kcna1-null mice separate from seizure control, and suggest that this widely used peptide may exert direct nootropic effects in the epileptic brain.
Collapse
|
103
|
Soloaga A, Pueta M, Cruz FB, Kembro JM, Marin RH. Chronic stress in Lizards: Studies on the Behavior and Benzodiazepine Receptors in Liolaemus koslowskyi and Cnemidophorus tergolaevigatus. ACTA ACUST UNITED AC 2017; 325:713-725. [PMID: 28198153 DOI: 10.1002/jez.2063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 01/09/2017] [Accepted: 01/10/2017] [Indexed: 12/12/2022]
Abstract
Behavioral and physiological adaptive responses of animals facing chronic exposure to a single stressor may allow them to overcome its negative effects for future exposures to similar stressful situations. At chemical level, the GABAA /benzodiazepine complex is considered one of the main receptor systems involved in the modulation of stress-induced responses. Here, we describe the behavioral responses of two different lizard species, Liolaemus koslowskyi and Cnemidophorus tergolaevigatus exposed to three potential chronic stressful treatments: (a) high temperature, (b) forced swimming, and (c) simulated predator. Additionally, we aimed to determine in those lizards whether the central-type benzodiazepine receptor (CBR; an allosteric modulator site of the GABAA receptor) is related to adaptive responses to those stressful stimulations. Our results revealed that the simulated predator was the stress condition that showed the largest difference in behavioral responses between the two species, resembling previously described strategies in nature. The basal affinity of CBRs (obtained from undisturbed animals) showed differences between both species, and the simulated predator was the only stressor that altered the affinity of CBRs. L. koslowskyi CBRs showed a decreased receptor affinity, whereas C. tergolaevigatus showed an increased receptor affinity in comparison to their respective control groups. We show for the first time the effects of different types of stressors upon behavioral responses and CBR biochemical parameters in two lizard species. Our findings suggest a potential GABA/benzodiazepine role in the ability of lizards to cope with a repeated exposure to a stressful (e.g., predator) condition.
Collapse
Affiliation(s)
- Alejandra Soloaga
- Centro Regional de Investigaciones Científicas y Transferencia Tecnológica, La Rioja (CONICET), Anillaco, La Rioja, Argentina
| | - Mariana Pueta
- Laboratorio de Fotobiología, Instituto de Investigaciones en Biodiversidad y Medioambiente (CONICET-UNComa), San Carlos de Bariloche, Río Negro, Argentina.,Departamento de Biología General, Centro Regional Universitario Bariloche, Universidad Nacional del Comahue, San Carlos de Bariloche, Río Negro, Argentina
| | - Félix Benjamín Cruz
- Laboratorio de Fotobiología, Instituto de Investigaciones en Biodiversidad y Medioambiente (CONICET-UNComa), San Carlos de Bariloche, Río Negro, Argentina
| | - Jackelyn Melissa Kembro
- Instituto de Investigaciones Biológicas y Tecnológicas (IIByT, CONICET-FCEFyN-UNC) and Instituto de Ciencia y Tecnología de los alimentos (ICTA), Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Raul Hector Marin
- Instituto de Investigaciones Biológicas y Tecnológicas (IIByT, CONICET-FCEFyN-UNC) and Instituto de Ciencia y Tecnología de los alimentos (ICTA), Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
104
|
Abstract
In this article, I describe how the current practice of classifying as a stressor any event that is accompanied by a change in any of a number of biological or behavioral measures-even when it is not accompanied by a long-term compromise in an organism's health or capacity to cope with daily challenges-has limited the utility of this concept. This permissive posture, which began with Selye's writings more than 65 years ago, is sustained by the public's desire for a simple term that might explain the tension generated by the threat of terrorists, growing economic inequality, increased competiveness in the workplace or for admission to the best universities, rogue nuclear bombs, and media reports of threats to health in food and water. I believe that the concept stress should be limited to select events that pose a serious threat to an organism's well-being or discarded as too ambiguous to be theoretically useful.
Collapse
|
105
|
Cissé M, Duplan E, Checler F. The transcription factor XBP1 in memory and cognition: Implications in Alzheimer disease. Mol Med 2017; 22:905-917. [PMID: 28079229 DOI: 10.2119/molmed.2016.00229] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 12/23/2016] [Indexed: 12/21/2022] Open
Abstract
X-box binding protein 1 (XBP1) is a unique basic region leucine zipper transcription factor isolated two decades ago in a search for regulators of major histocompatibility complex class II gene expression. XBP1 is a very complex protein regulating many physiological functions, including immune system, inflammatory responses, and lipid metabolism. Evidence over the past few years suggests that XBP1 also plays important roles in pathological settings since its activity as transcription factor has profound effects on the prognosis and progression of diseases such as cancer, neurodegeneration, and diabetes. Here we provide an overview on recent advances in our understanding of this multifaceted molecule, particularly in regulating synaptic plasticity and memory function, and the implications in neurodegenerative diseases with emphasis on Alzheimer disease.
Collapse
Affiliation(s)
- Moustapha Cissé
- Université Côte d'Azur, INSERM, CNRS, IPMC, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| | - Eric Duplan
- Université Côte d'Azur, INSERM, CNRS, IPMC, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| | - Frédéric Checler
- Université Côte d'Azur, INSERM, CNRS, IPMC, team labeled "Fondation pour la Recherche Médicale" and "Laboratory of Excellence (LABEX) Distalz", 660 route des Lucioles, 06560, Sophia-Antipolis, Valbonne, France
| |
Collapse
|
106
|
Wang JH, Lei X, Cheng XR, Zhang XR, Liu G, Cheng JP, Xu YR, Zeng J, Zhou WX, Zhang YX. LW-AFC, a new formula derived from Liuwei Dihuang decoction, ameliorates behavioral and pathological deterioration via modulating the neuroendocrine-immune system in PrP-hAβPPswe/PS1 ΔE9 transgenic mice. Alzheimers Res Ther 2016; 8:57. [PMID: 27964740 PMCID: PMC5154149 DOI: 10.1186/s13195-016-0226-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 11/24/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Accumulating evidence implicates the neuroendocrine immunomodulation (NIM) network in the physiopathological mechanism of Alzheimer's disease (AD). Notably, we previously revealed that the NIM network is dysregulated in the PrP-hAβPPswe/PS1ΔE9 (APP/PS1) transgenic mouse model of AD. METHODS After treatment with a novel Liuwei Dihuang formula (LW-AFC), mice were cognitively evaluated in behavioral experiments. Neuron loss, amyloid-β (Aβ) deposition, and Aβ level were analyzed using Nissl staining, immunofluorescence, and an AlphaLISA assay, respectively. Multiplex bead analysis, a radioimmunoassay, immunochemiluminometry, and an enzyme-linked immunosorbent assay (ELISA) were used to measure cytokine and hormone levels. Lymphocyte subsets were detected using flow cytometry. Data between two groups were compared using a Student's t test. Comparison of the data from multiple groups against one group was performed using a one-way analysis of variance (ANOVA) followed by a Dunnett's post hoc test or a two-way repeated-measures analysis of variance with a Tukey multiple comparisons test. RESULTS LW-AFC ameliorated the cognitive impairment observed in APP/PS1 mice, including the impairment of object recognition memory, spatial learning and memory, and active and passive avoidance. In addition, LW-AFC alleviated the neuron loss in the hippocampus, suppressed Aβ deposition in the brain, and reduced the concentration of Aβ1-42 in the hippocampus and plasma of APP/PS1 mice. LW-AFC treatment also significantly decreased the secretion of corticotropin-releasing hormone and gonadotropin-releasing hormone in the hypothalamus, and adrenocorticotropic hormone, luteinizing hormone, and follicle-stimulating hormone in the pituitary. Moreover, LW-AFC increased CD8+CD28+ T cells, and reduced CD4+CD25+Foxp3+ T cells in the spleen lymphocytes, downregulated interleukin (IL)-1β, IL-2, IL-6, IL-23, granulocyte-macrophage colony stimulating factor, and tumor necrosis factor-α and -β, and upregulated IL-4 and granulocyte colony stimulating factor in the plasma of APP/PS1 mice. CONCLUSIONS LW-AFC ameliorated the behavioral and pathological deterioration of APP/PS1 transgenic mice via the restoration of the NIM network to a greater extent than either memantine or donepezil, which supports the use of LW-AFC as a potential agent for AD therapy.
Collapse
Affiliation(s)
- Jian-Hui Wang
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Xi Lei
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
- Guangxi Medical University, Nanning, 530021, China
| | - Xiao-Rui Cheng
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China.
| | - Xiao-Rui Zhang
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Gang Liu
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Jun-Ping Cheng
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Yi-Ran Xu
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Ju Zeng
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Wen-Xia Zhou
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China.
| | - Yong-Xiang Zhang
- Department of Neuroimmunopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China.
| |
Collapse
|
107
|
ProBDNF Signaling Regulates Depression-Like Behaviors in Rodents under Chronic Stress. Neuropsychopharmacology 2016; 41:2882-2892. [PMID: 27312407 PMCID: PMC5061880 DOI: 10.1038/npp.2016.100] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 06/09/2016] [Accepted: 06/12/2016] [Indexed: 01/08/2023]
Abstract
Chronic exposure to stressful environment is a key risk factor contributing to the development of depression. However, the mechanisms involved in this process are still unclear. Brain-derived neurotropic factor (BDNF) has long been investigated for its positive role in regulation of mood, although the role of its precursor, proBDNF, in regulation of mood is not known. In this study, using an unpredictable chronic mild stress (UCMS) paradigm we found that the protein levels of proBDNF were increased in the neocortex and hippocampus of stressed mice and this UCMS-induced upregulation of proBDNF was abolished by chronic administration of fluoxetine. We then established a rat model of UCMS and found that the expression of proBDNF/p75NTR/sortilin was upregulated, whereas the expression of mature BDNF and TrkB was downregulated in both neocortex and hippocampus of chronically stressed rats. Finally, we found that the injection of anti-proBDNF antibody via intracerebroventricular (i.c.v.) and intraperitoneal (i.p.) approaches into the UCMS rats significantly reversed the stress-induced depression-like behavior and restored the exploratory activity and spine growth. Although intramuscular injection of AAV-proBDNF did not exacerbate the UCMS-elicited rat mood-related behavioral or pathological abnormalities, i.c.v. injection of AAV-proBDNF increased the depression-like behavior in naive rats. Our findings suggest that proBDNF plays a role in the development of chronic stress-induced mood disturbances in rodents. Central (i.c.v.) or peripheral (i.p.) inhibition of proBDNF by injecting specific anti-proBDNF antibodies may provide a novel therapeutic approach for the treatment of stress-related mood disorders.
Collapse
|
108
|
Arango-Lievano M, Jeanneteau F. Timing and crosstalk of glucocorticoid signaling with cytokines, neurotransmitters and growth factors. Pharmacol Res 2016; 113:1-17. [DOI: 10.1016/j.phrs.2016.08.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 08/02/2016] [Accepted: 08/02/2016] [Indexed: 01/05/2023]
|
109
|
Molet J, Maras PM, Kinney-Lang E, Harris NG, Rashid F, Ivy AS, Solodkin A, Obenaus A, Baram TZ. MRI uncovers disrupted hippocampal microstructure that underlies memory impairments after early-life adversity. Hippocampus 2016; 26:1618-1632. [PMID: 27657911 DOI: 10.1002/hipo.22661] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2016] [Indexed: 12/13/2022]
Abstract
Memory and related cognitive functions are progressively impaired in a subgroup of individuals experiencing childhood adversity and stress. However, it is not possible to identify vulnerable individuals early, a crucial step for intervention. In this study, high-resolution magnetic resonance imaging (MRI) and intra-hippocampal diffusion tensor imaging (DTI) were employed to examine for structural signatures of cognitive adolescent vulnerabilities in a rodent model of early-life adversity. These methods were complemented by neuroanatomical and functional assessments of hippocampal network integrity during adolescence, adulthood and middle-age. The high-resolution MRI identified selective loss of dorsal hippocampal volume, and intra-hippocampal DTI uncovered disruption of dendritic structure, consistent with disrupted local connectivity, already during late adolescence in adversity-experiencing rats. Memory deteriorated over time, and stunting of hippocampal dendritic trees was apparent on neuroanatomical analyses. Thus, disrupted hippocampal neuronal structure and connectivity, associated with cognitive impairments, are detectable via non-invasive imaging modalities in rats experiencing early-life adversity. These high-resolution imaging approaches may constitute promising tools for prediction and assessment of at-risk individuals in the clinic. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Pamela M Maras
- Department of Pediatrics, UC-Irvine, Irvine, CA, 92697-4475, USA
| | - Eli Kinney-Lang
- Department of Pediatrics, UC-Irvine, Irvine, CA, 92697-4475, USA.,Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Neil G Harris
- Department of Neurosurgery, UCLA, Los Angeles, CA, 90095-6901, USA
| | - Faisal Rashid
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | | | - Ana Solodkin
- Department of Anatomy and Neurobiology.,Department of Neurology, UC-Irvine, Irvine, CA, 92697-4475, USA
| | - Andre Obenaus
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Tallie Z Baram
- Department of Anatomy and Neurobiology.,Department of Pediatrics, UC-Irvine, Irvine, CA, 92697-4475, USA.,Department of Neurology, UC-Irvine, Irvine, CA, 92697-4475, USA
| |
Collapse
|
110
|
Rahman MM, Callaghan CK, Kerskens CM, Chattarji S, O'Mara SM. Early hippocampal volume loss as a marker of eventual memory deficits caused by repeated stress. Sci Rep 2016; 6:29127. [PMID: 27374165 PMCID: PMC4931588 DOI: 10.1038/srep29127] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 06/15/2016] [Indexed: 02/07/2023] Open
Abstract
Exposure to severe and prolonged stress has detrimental effects on the hippocampus. However, relatively little is known about the gradual changes in hippocampal structure, and its behavioral consequences, over the course of repeated stress. Behavioral analyses during 10 days of chronic stress pointed to a delayed decline in spatial memory, the full impact of which is evident only after the end of stress. In contrast, concurrent volumetric measurements in the same animals revealed significant reduction in hippocampal volumes in stressed animals relative to their unstressed counterparts, as early as the third day of stress. Notably, animals that were behaviorally the worst affected at the end of chronic stress suffered the most pronounced early loss in hippocampal volume. Together, these findings support the view that not only is smaller hippocampal volume linked to stress-induced memory deficits, but it may also act as an early risk factor for the eventual development of cognitive impairments seen in stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Mohammed Mostafizur Rahman
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India.,Institute of Neuroscience, Trinity College Dublin, College Green, Dublin 2, Ireland
| | | | - Christian M Kerskens
- Institute of Neuroscience, Trinity College Dublin, College Green, Dublin 2, Ireland
| | - Sumantra Chattarji
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore 560065, India
| | - Shane M O'Mara
- Institute of Neuroscience, Trinity College Dublin, College Green, Dublin 2, Ireland
| |
Collapse
|
111
|
White SL, Vassoler FM, Schmidt HD, Pierce RC, Wimmer ME. Enhanced anxiety in the male offspring of sires that self-administered cocaine. Addict Biol 2016; 21:802-810. [PMID: 25923597 DOI: 10.1111/adb.12258] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We previously showed that paternal cocaine exposure reduced the reinforcing efficacy of cocaine in male offspring. Here, we sought to determine whether paternal cocaine experience could also influence anxiety levels in offspring. Male rats were allowed to self-administer cocaine (controls received saline passively) for 60 days and then were bred with naïve females. Measures of anxiety and cocaine-induced anxiogenic effects were assessed in the adult offspring. Cocaine-sired male offspring exhibited increased anxiety-like behaviors, as measured using the novelty-induced hypophagia and defensive burying tasks, relative to saline-sired males. In contrast, sire cocaine experience had no effect on anxiety-like behaviors in female offspring. When challenged with an anxiogenic (but not anorectic) dose of cocaine (2.5 mg/kg, i.p.), anxiety-like behavior was enhanced in all animals to an equal degree regardless of sire drug experience. Since anxiety and depression are often co-morbid, we also assessed measures of depressive-like behavior. Sire cocaine experience had no effect on depression-like behaviors, as measured by the forced swim task, among male offspring. In a separate group of naïve littermates, select neuronal correlates of anxiety were measured. Male offspring of cocaine-experienced sires showed increased mRNA and protein expression of corticotropin-releasing factor receptor 2 in the hippocampus. Together, these results indicate that cocaine-experienced sires produce male progeny that have increased baseline anxiety, which is unaltered by subsequent cocaine exposure.
Collapse
Affiliation(s)
- Samantha L. White
- Center for Neurobiology and Behavior; Department of Psychiatry; Perelman School of Medicine; University of Pennsylvania; Philadelphia PA USA
| | - Fair M. Vassoler
- Center for Neurobiology and Behavior; Department of Psychiatry; Perelman School of Medicine; University of Pennsylvania; Philadelphia PA USA
| | - Heath D. Schmidt
- Center for Neurobiology and Behavior; Department of Psychiatry; Perelman School of Medicine; University of Pennsylvania; Philadelphia PA USA
| | - R. Christopher Pierce
- Center for Neurobiology and Behavior; Department of Psychiatry; Perelman School of Medicine; University of Pennsylvania; Philadelphia PA USA
| | - Mathieu E. Wimmer
- Center for Neurobiology and Behavior; Department of Psychiatry; Perelman School of Medicine; University of Pennsylvania; Philadelphia PA USA
| |
Collapse
|
112
|
Chmielewski NN, Caressi C, Giedzinski E, Parihar VK, Limoli CL. Contrasting the effects of proton irradiation on dendritic complexity of subiculum neurons in wild type and MCAT mice. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2016; 57:364-371. [PMID: 26996825 DOI: 10.1002/em.22006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 02/19/2016] [Indexed: 06/05/2023]
Abstract
Growing evidence suggests that radiation-induced oxidative stress directly affects a wide range of biological changes with an overall negative impact on CNS function. In the past we have demonstrated that transgenic mice over-expressing human catalase targeted to the mitochondria (MCAT) exhibit a range of neuroprotective phenotypes following irradiation that include improved neurogenesis, dendritic complexity, and cognition. To determine the extent of the neuroprotective phenotype afforded by MCAT expression in different hippocampal regions, we analyzed subiculum neurons for changes in neuronal structure and synaptic integrity after exposure to low dose (0.5 Gy) 150 MeV proton irradiation. One month following irradiation of WT and MCAT mice, a range of morphometric parameters were quantified along Golgi-Cox impregnated neurons. Compared with WT mice, subiculum neurons from MCAT mice exhibited increased trends (albeit not statistically significant) toward increased dendritic complexity in both control and irradiated cohorts. However, Sholl analysis of MCAT mice revealed significantly increased arborization of the distal dendritic tree, indicating a protective effect on secondary and tertiary branching. Interestingly, radiation-induced increases in postsynaptic density protein (PSD-95) puncta were not as pronounced in MCAT compared with WT mice, and were significantly lower after the 0.5 Gy dose. As past data has linked radiation exposure to reduced dendritic complexity, elevated PSD-95 and impaired cognition, reductions in mitochondrial oxidative stress have proven useful in ameliorating many of these radiation-induced sequelae. Data presented here shows similar trends, and again points to the potential benefits of reducing oxidative stress in the brain to attenuate radiation injury. Environ. Mol. Mutagen. 57:364-371, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Chongshan Caressi
- Department of Radiation Oncology, University of California, Irvine, California
| | - Erich Giedzinski
- Department of Radiation Oncology, University of California, Irvine, California
| | - Vipan K Parihar
- Department of Radiation Oncology, University of California, Irvine, California
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, California
| |
Collapse
|
113
|
Howland MA, Sandman CA, Glynn LM, Crippen C, Davis EP. Fetal exposure to placental corticotropin-releasing hormone is associated with child self-reported internalizing symptoms. Psychoneuroendocrinology 2016; 67:10-7. [PMID: 26855003 PMCID: PMC4808336 DOI: 10.1016/j.psyneuen.2016.01.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 01/21/2016] [Accepted: 01/25/2016] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Fetal exposure to maternal prenatal stress hormones such as cortisol exerts influences on the developing nervous system that persist and include risk for internalizing symptoms later in life. Placental corticotropin-releasing hormone (pCRH) is a feto-placental stress signal that also shapes fetal neurodevelopment and may be a more direct indicator of the fetal experience than maternal stress hormones. The programming effects of pCRH on child development are unknown. The current investigation examined associations between prenatal maternal and placental stress hormone exposures (maternal cortisol and pCRH) and child self-reported internalizing symptoms at age 5. METHOD Maternal plasma cortisol and pCRH levels were measured at 15, 19, 25, 31, and 36 weeks' gestation in a sample of 83 women and their 91 children (8 sibling pairs from separate pregnancies), who were born full-term. Child self-reported internalizing symptoms at age 5 were obtained using scales of the Berkeley Puppet Interview. RESULTS Placental CRH profiles (including elevations in mid-gestation) were associated with higher levels of internalizing symptoms at age 5. This effect was not explained by critical prenatal or postnatal influences, including obstetric risk, concurrent maternal psychological state, and family socio-economic status. Prenatal maternal cortisol was not significantly associated with child self-reported internalizing symptoms. CONCLUSIONS Findings suggest that elevated exposures to the feto-placental stress signal pCRH exert programming effects on the developing fetal central nervous system, with lasting consequences for child mental health.
Collapse
|
114
|
Su G, Zhao T, Zhao Y, Sun-Waterhouse D, Qiu C, Huang P, Zhao M. Effect of anchovy (Coilia mystus) protein hydrolysate and its Maillard reaction product on combating memory-impairment in mice. Food Res Int 2016. [DOI: 10.1016/j.foodres.2016.01.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
115
|
Stress Response and Perinatal Reprogramming: Unraveling (Mal)adaptive Strategies. Neural Plast 2016; 2016:6752193. [PMID: 27057367 PMCID: PMC4812483 DOI: 10.1155/2016/6752193] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/17/2016] [Indexed: 12/28/2022] Open
Abstract
Environmental stressors induce coping strategies in the majority of individuals. The stress response, involving the activation of the hypothalamic-pituitary-adrenocortical axis and the consequent release of corticosteroid hormones, is indeed aimed at promoting metabolic, functional, and behavioral adaptations. However, behavioral stress is also associated with fast and long-lasting neurochemical, structural, and behavioral changes, leading to long-term remodeling of glutamate transmission, and increased susceptibility to neuropsychiatric disorders. Of note, early-life events, both in utero and during the early postnatal life, trigger reprogramming of the stress response, which is often associated with loss of stress resilience and ensuing neurobehavioral (mal)adaptations. Indeed, adverse experiences in early life are known to induce long-term stress-related neuropsychiatric disorders in vulnerable individuals. Here, we discuss recent findings about stress remodeling of excitatory neurotransmission and brain morphology in animal models of behavioral stress. These changes are likely driven by epigenetic factors that lie at the core of the stress-response reprogramming in individuals with a history of perinatal stress. We propose that reprogramming mechanisms may underlie the reorganization of excitatory neurotransmission in the short- and long-term response to stressful stimuli.
Collapse
|
116
|
Zheng Y, Zhang YM, Ni X. Urocortin 2 But Not Urocortin 3 Promotes the Synaptic Formation in Hipppocampal Neurons via Induction of NGF Production by Astrocytes. Endocrinology 2016; 157:1200-10. [PMID: 26713785 DOI: 10.1210/en.2015-1812] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CRH family peptides play differential role during various physiological and pathophysiological responses, such as stress. Urocortins (UCNs) have been implicated to play complementary or contrasting actions for the effects of CRH during stress. It has been shown that activation of CRH receptor type 1 (CRHR1) results in decreased synapse formation in hippocampus. We therefore explored the effect of UCN2 and UCN3, the exclusive CRHR2 agonists, on synaptic formation in hippocampus. In hippocampal slices cultures, UCN2 but not UCN3 treatment increased the levels of presynaptic protein synapsinI and postsynaptic protein postsynaptic density 95 (PSD95), which was reversed by CRHR2 antagonist astressin 2B. In isolated hippocampal neurons, however, UCN2 decreased the numbers of synapsinI- and PSD95-labeled terminals/clusters via CRHR2. Treatment of hippocampal neurons with the media of UCN2-treated astrocytes led to an increase in synapsinI- and PSD95-labeled terminals. In neuron-astrocyte cocultures, UCN2 also enhanced the numbers and level of synapsinI- and PSD95-labeled terminals. These effects did not occur if glial cells were transfected with CRHR2 small interfering RNA. UCN2 but not UCN3 treatment induced nerve growth factor (NGF) production in astrocytes via CRHR2. The effects of the media of UCN2-treated glial cells on synapse formation in hippocampal neurons were prevented by administration of NGF receptor antagonists. Our data indicate that UCN2 promotes synapse formation in hippocampus via induction of NGF secretion from astrocytes. CRHR2 in glial cells mediates the stimulatory effects of CRH. Glia-neuron communication is critical for neuronal circuits remodeling and synaptic plasticity in response to neurohormones or neuromodulators.
Collapse
Affiliation(s)
- You Zheng
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| | - Yan-Min Zhang
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| | - Xin Ni
- Department of Physiology, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
117
|
van der Kooij MA, Masana M, Rust MB, Müller MB. The stressed cytoskeleton: How actin dynamics can shape stress-related consequences on synaptic plasticity and complex behavior. Neurosci Biobehav Rev 2016; 62:69-75. [DOI: 10.1016/j.neubiorev.2015.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 12/01/2015] [Accepted: 12/07/2015] [Indexed: 01/06/2023]
|
118
|
PSD-95 regulates CRFR1 localization, trafficking and β-arrestin2 recruitment. Cell Signal 2016; 28:531-540. [PMID: 26898829 DOI: 10.1016/j.cellsig.2016.02.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 02/16/2016] [Accepted: 02/16/2016] [Indexed: 12/13/2022]
Abstract
Corticotropin-releasing factor (CRF) is a neuropeptide commonly associated with the hypothalamic-pituitary adrenal axis stress response. Upon release, CRF activates two G protein-coupled receptors (GPCRs): CRF receptor 1 (CRFR1) and CRF receptor 2 (CRFR2). Although both receptors contribute to mood regulation, CRFR1 antagonists have demonstrated anxiolytic and antidepressant-like properties that may be exploited in the generation of new pharmacological interventions for mental illnesses. Previous studies have demonstrated CRFR1 capable of heterologously sensitizing serotonin 2A receptor (5-HT2AR) signaling: another GPCR implicated in psychiatric disease. Interestingly, this phenomenon was dependent on Postsynaptic density 95 (PSD-95)/Disc Large/Zona Occludens (PDZ) interactions on the distal carboxyl termini of both receptors. In the current study, we demonstrate that endogenous PSD-95 can be co-immunoprecipitated with CRFR1 from cortical brain homogenate, and this interaction appears to be primarily via the PDZ-binding motif. Additionally, PSD-95 colocalizes with CRFR1 within the dendritic projections of cultured mouse neurons in a PDZ-binding motif-dependent manner. In HEK 293 cells, PSD-95 overexpression inhibited CRFR1 endocytosis, whereas PSD-95 shRNA knockdown enhanced CRFR1 endocytosis. Although PSD-95 does not appear to play a significant role in CRF-mediated cAMP or ERK1/2 signaling, PSD-95 was demonstrated to suppress β-arrestin2 recruitment: providing a potential mechanism for PSD-95's inhibition of endocytosis. In revisiting previously documented heterologous sensitization, PSD-95 shRNA knockdown did not prevent CRFR1-mediated enhancement of 5-HT2AR signaling. In conclusion, we have identified and characterized a novel functional relationship between CRFR1 and PSD-95 that may have implications in the design of new treatment strategies for mental illness.
Collapse
|
119
|
Chen A. Genetic Dissection of the Neuroendocrine and Behavioral Responses to Stressful Challenges. STEM CELLS IN NEUROENDOCRINOLOGY 2016. [DOI: 10.1007/978-3-319-41603-8_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
120
|
Chattarji S, Tomar A, Suvrathan A, Ghosh S, Rahman MM. Neighborhood matters: divergent patterns of stress-induced plasticity across the brain. Nat Neurosci 2015; 18:1364-75. [PMID: 26404711 DOI: 10.1038/nn.4115] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/19/2015] [Indexed: 02/07/2023]
Abstract
The fact that exposure to severe stress leads to the development of psychiatric disorders serves as the basic rationale for animal models of stress disorders. Clinical and neuroimaging studies have shown that three brain areas involved in learning and memory--the hippocampus, amygdala and prefrontal cortex--undergo distinct structural and functional changes in individuals with stress disorders. These findings from patient studies pose several challenges for animal models of stress disorders. For instance, why does stress impair cognitive function, yet enhance fear and anxiety? Can the same stressful experience elicit contrasting patterns of plasticity in the hippocampus, amygdala and prefrontal cortex? How does even a brief exposure to traumatic stress lead to long-lasting behavioral abnormalities? Thus, animal models of stress disorders must not only capture the unique spatio-temporal features of structural and functional alterations in these brain areas, but must also provide insights into the underlying neuronal plasticity mechanisms. This Review will address some of these key questions by describing findings from animal models on how stress-induced plasticity varies across different brain regions and thereby gives rise to the debilitating emotional and cognitive symptoms of stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Sumantra Chattarji
- Centre for Brain Development and Repair, Institute of Stem Cell Biology and Regenerative Medicine, National Centre for Biological Sciences, Bangalore, India
| | - Anupratap Tomar
- Laboratory for Circuit and Behavioral Physiology, RIKEN Brain Science Institute, Wakoshi, Saitama, Japan
| | - Aparna Suvrathan
- Department of Neurobiology, Stanford University, Stanford, California, USA
| | - Supriya Ghosh
- Department of Neurobiology, University of Chicago, Chicago, Illinois, USA
| | - Mohammed Mostafizur Rahman
- Centre for Brain Development and Repair, Institute of Stem Cell Biology and Regenerative Medicine, National Centre for Biological Sciences, Bangalore, India
| |
Collapse
|
121
|
McFalls AJ, Imperio CG, Bixler G, Freeman WM, Grigson PS, Vrana KE. Reward devaluation and heroin escalation is associated with differential expression of CRF signaling genes. Brain Res Bull 2015; 123:81-93. [PMID: 26655889 DOI: 10.1016/j.brainresbull.2015.11.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/04/2015] [Accepted: 11/05/2015] [Indexed: 01/08/2023]
Abstract
One of the most damaging aspects of drug addiction is the degree to which natural rewards (family, friends, employment) are devalued in favor of seeking, obtaining and taking drugs. We have utilized an animal model of reward devaluation and heroin self-administration to explore the role of the coricotropin releasing factor (CRF) pathway. Given access to a saccharin cue followed by the opportunity to self-administer heroin, animals will parse into distinct phenotypes that suppress their saccharin intake (in favor of escalating heroin self-administration) or vice versa. We find that large saccharin suppressors (large heroin takers) demonstrate increased mRNA expression for elements of the CRF signaling pathway (CRF, CRF receptors and CRF binding protein) within the hippocampus, medial prefrontal cortex and the ventral tegmental area. Moreover, there were no gene expression changes of these components in the nucleus accumbens. Use of bisulfite conversion sequencing suggests that changes in CRF binding protein and CRF receptor gene expression may be mediated by differential promoter methylation.
Collapse
Affiliation(s)
- Ashley J McFalls
- Departments of Pharmacology, Penn State College of Medicine, Hershey, PA, United States
| | - Caesar G Imperio
- Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States
| | - Georgina Bixler
- Departments of Pharmacology, Penn State College of Medicine, Hershey, PA, United States
| | - Willard M Freeman
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Patricia Sue Grigson
- Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States
| | - Kent E Vrana
- Departments of Pharmacology, Penn State College of Medicine, Hershey, PA, United States.
| |
Collapse
|
122
|
Sooy K, Noble J, McBride A, Binnie M, Yau JLW, Seckl JR, Walker BR, Webster SP. Cognitive and Disease-Modifying Effects of 11β-Hydroxysteroid Dehydrogenase Type 1 Inhibition in Male Tg2576 Mice, a Model of Alzheimer's Disease. Endocrinology 2015; 156:4592-603. [PMID: 26305888 PMCID: PMC4655221 DOI: 10.1210/en.2015-1395] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Chronic exposure to elevated levels of glucocorticoids has been linked to age-related cognitive decline and may play a role in Alzheimer's disease. In the brain, 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1) amplifies intracellular glucocorticoid levels. We show that short-term treatment of aged, cognitively impaired C57BL/6 mice with the potent and selective 11β-HSD1 inhibitor UE2316 improves memory, including after intracerebroventricular drug administration to the central nervous system alone. In the Tg2576 mouse model of Alzheimer's disease, UE2316 treatment of mice aged 14 months for 4 weeks also decreased the number of β-amyloid (Aβ) plaques in the cerebral cortex, associated with a selective increase in local insulin-degrading enzyme (involved in Aβ breakdown and known to be glucocorticoid regulated). Chronic treatment of young Tg2576 mice with UE2316 for up to 13 months prevented cognitive decline but did not prevent Aβ plaque formation. We conclude that reducing glucocorticoid regeneration in the brain improves cognition independently of reduced Aβ plaque pathology and that 11β-HSD1 inhibitors have potential as cognitive enhancers in age-associated memory impairment and Alzheimer's dementia.
Collapse
Affiliation(s)
- Karen Sooy
- University/BHF Centre for Cardiovascular Science (K.S., J.N., A.M., M.B., J.L.W.Y., J.R.S., B.R.W., S.P.W.), Queen's Medical Research Institute, and Centre for Cognitive Aging and Cognitive Epidemiology (J.L.W.Y., J.R.S.), University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - June Noble
- University/BHF Centre for Cardiovascular Science (K.S., J.N., A.M., M.B., J.L.W.Y., J.R.S., B.R.W., S.P.W.), Queen's Medical Research Institute, and Centre for Cognitive Aging and Cognitive Epidemiology (J.L.W.Y., J.R.S.), University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Andrew McBride
- University/BHF Centre for Cardiovascular Science (K.S., J.N., A.M., M.B., J.L.W.Y., J.R.S., B.R.W., S.P.W.), Queen's Medical Research Institute, and Centre for Cognitive Aging and Cognitive Epidemiology (J.L.W.Y., J.R.S.), University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Margaret Binnie
- University/BHF Centre for Cardiovascular Science (K.S., J.N., A.M., M.B., J.L.W.Y., J.R.S., B.R.W., S.P.W.), Queen's Medical Research Institute, and Centre for Cognitive Aging and Cognitive Epidemiology (J.L.W.Y., J.R.S.), University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Joyce L W Yau
- University/BHF Centre for Cardiovascular Science (K.S., J.N., A.M., M.B., J.L.W.Y., J.R.S., B.R.W., S.P.W.), Queen's Medical Research Institute, and Centre for Cognitive Aging and Cognitive Epidemiology (J.L.W.Y., J.R.S.), University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Jonathan R Seckl
- University/BHF Centre for Cardiovascular Science (K.S., J.N., A.M., M.B., J.L.W.Y., J.R.S., B.R.W., S.P.W.), Queen's Medical Research Institute, and Centre for Cognitive Aging and Cognitive Epidemiology (J.L.W.Y., J.R.S.), University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Brian R Walker
- University/BHF Centre for Cardiovascular Science (K.S., J.N., A.M., M.B., J.L.W.Y., J.R.S., B.R.W., S.P.W.), Queen's Medical Research Institute, and Centre for Cognitive Aging and Cognitive Epidemiology (J.L.W.Y., J.R.S.), University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Scott P Webster
- University/BHF Centre for Cardiovascular Science (K.S., J.N., A.M., M.B., J.L.W.Y., J.R.S., B.R.W., S.P.W.), Queen's Medical Research Institute, and Centre for Cognitive Aging and Cognitive Epidemiology (J.L.W.Y., J.R.S.), University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| |
Collapse
|
123
|
Chen Y, Molet J, Gunn BG, Ressler K, Baram TZ. Diversity of Reporter Expression Patterns in Transgenic Mouse Lines Targeting Corticotropin-Releasing Hormone-Expressing Neurons. Endocrinology 2015; 156:4769-80. [PMID: 26402844 PMCID: PMC4655217 DOI: 10.1210/en.2015-1673] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Transgenic mice, including lines targeting corticotropin-releasing factor (CRF or CRH), have been extensively employed to study stress neurobiology. These powerful tools are poised to revolutionize our understanding of the localization and connectivity of CRH-expressing neurons, and the crucial roles of CRH in normal and pathological conditions. Accurate interpretation of studies using cell type-specific transgenic mice vitally depends on congruence between expression of the endogenous peptide and reporter. If reporter expression does not faithfully reproduce native gene expression, then effects of manipulating unintentionally targeted cells may be misattributed. Here, we studied CRH and reporter expression patterns in 3 adult transgenic mice: Crh-IRES-Cre;Ai14 (tdTomato mouse), Crfp3.0CreGFP, and Crh-GFP BAC. We employed the CRH antiserum generated by Vale after validating its specificity using CRH-null mice. We focused the analyses on stress-salient regions, including hypothalamus, amygdala, bed nucleus of the stria terminalis, and hippocampus. Expression patterns of endogenous CRH were consistent among wild-type and transgenic mice. In tdTomato mice, most CRH-expressing neurons coexpressed the reporter, yet the reporter identified a few non-CRH-expressing pyramidal-like cells in hippocampal CA1 and CA3. In Crfp3.0CreGFP mice, coexpression of CRH and the reporter was found in central amygdala and, less commonly, in other evaluated regions. In Crh-GFP BAC mice, the large majority of neurons expressed either CRH or reporter, with little overlap. These data highlight significant diversity in concordant expression of reporter and endogenous CRH among 3 available transgenic mice. These findings should be instrumental in interpreting important scientific findings emerging from the use of these potent neurobiological tools.
Collapse
Affiliation(s)
- Yuncai Chen
- Departments of Pediatrics (Y.C., B.G.G., T.Z.B.) and Anatomy/Neurobiology (Y.C., J.M., B.G.G., T.Z.B.), University of California, Irvine, Irvine, California 92697-4475; and Department of Psychiatry and Behavioral Sciences (K.R.), Emory University, Atlanta, Georgia 30322-4250
| | - Jenny Molet
- Departments of Pediatrics (Y.C., B.G.G., T.Z.B.) and Anatomy/Neurobiology (Y.C., J.M., B.G.G., T.Z.B.), University of California, Irvine, Irvine, California 92697-4475; and Department of Psychiatry and Behavioral Sciences (K.R.), Emory University, Atlanta, Georgia 30322-4250
| | - Benjamin G Gunn
- Departments of Pediatrics (Y.C., B.G.G., T.Z.B.) and Anatomy/Neurobiology (Y.C., J.M., B.G.G., T.Z.B.), University of California, Irvine, Irvine, California 92697-4475; and Department of Psychiatry and Behavioral Sciences (K.R.), Emory University, Atlanta, Georgia 30322-4250
| | - Kerry Ressler
- Departments of Pediatrics (Y.C., B.G.G., T.Z.B.) and Anatomy/Neurobiology (Y.C., J.M., B.G.G., T.Z.B.), University of California, Irvine, Irvine, California 92697-4475; and Department of Psychiatry and Behavioral Sciences (K.R.), Emory University, Atlanta, Georgia 30322-4250
| | - Tallie Z Baram
- Departments of Pediatrics (Y.C., B.G.G., T.Z.B.) and Anatomy/Neurobiology (Y.C., J.M., B.G.G., T.Z.B.), University of California, Irvine, Irvine, California 92697-4475; and Department of Psychiatry and Behavioral Sciences (K.R.), Emory University, Atlanta, Georgia 30322-4250
| |
Collapse
|
124
|
Bangasser DA, Kawasumi Y. Cognitive disruptions in stress-related psychiatric disorders: A role for corticotropin releasing factor (CRF). Horm Behav 2015; 76:125-35. [PMID: 25888454 PMCID: PMC4605842 DOI: 10.1016/j.yhbeh.2015.04.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 04/06/2015] [Accepted: 04/07/2015] [Indexed: 12/28/2022]
Abstract
This article is part of a Special Issue "SBN 2014". Stress is a potential etiology contributor to both post-traumatic stress disorders (PTSD) and major depression. One stress-related neuropeptide that is hypersecreted in these disorders is corticotropin releasing factor (CRF). Dysregulation of CRF has long been linked to the emotion and mood symptoms that characterize PTSD and depression. However, the idea that CRF also mediates the cognitive disruptions observed in patients with these disorders has received less attention. Here we review literature indicating that CRF can alter cognitive functions. Detailed are anatomical studies revealing that CRF is poised to modulate regions required for learning and memory. We also describe preclinical behavioral studies that demonstrate CRF's ability to alter fear conditioning, impair memory consolidation, and alter a number of executive functions, including attention and cognitive flexibility. The implications of these findings for the etiology and treatment of the cognitive impairments observed in stress-related psychiatric disorders are described.
Collapse
Affiliation(s)
- Debra A Bangasser
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, USA.
| | - Yushi Kawasumi
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, USA
| |
Collapse
|
125
|
Bullmann T, Seeger G, Stieler J, Hanics J, Reimann K, Kretzschmann TP, Hilbrich I, Holzer M, Alpár A, Arendt T. Tau phosphorylation-associated spine regression does not impair hippocampal-dependent memory in hibernating golden hamsters. Hippocampus 2015; 26:301-18. [DOI: 10.1002/hipo.22522] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 08/12/2015] [Accepted: 08/31/2015] [Indexed: 01/12/2023]
Affiliation(s)
- Torsten Bullmann
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
- Frey Initiative Research Unit, RIKEN Quantitative Biology Center; Japan
| | - Gudrun Seeger
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Jens Stieler
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - János Hanics
- MTA-SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences; Budapest Hungary
- Department of Anatomy; Semmelweis University; Budapest Hungary
| | - Katja Reimann
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Tanja Petra Kretzschmann
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Isabel Hilbrich
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Max Holzer
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Alán Alpár
- MTA-SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences; Budapest Hungary
- Department of Anatomy; Semmelweis University; Budapest Hungary
| | - Thomas Arendt
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| |
Collapse
|
126
|
Kempf SJ, Sepe S, von Toerne C, Janik D, Neff F, Hauck SM, Atkinson MJ, Mastroberardino PG, Tapio S. Neonatal Irradiation Leads to Persistent Proteome Alterations Involved in Synaptic Plasticity in the Mouse Hippocampus and Cortex. J Proteome Res 2015; 14:4674-86. [PMID: 26420666 DOI: 10.1021/acs.jproteome.5b00564] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Recent epidemiological data indicate that radiation doses as low as those used in computer tomography may result in long-term neurocognitive side effects. The aim of this study was to elucidate long-term molecular alterations related to memory formation in the brain after low and moderate doses of γ radiation. Female C57BL/6J mice were irradiated on postnatal day 10 with total body doses of 0.1, 0.5, or 2.0 Gy; the control group was sham-irradiated. The proteome analysis of hippocampus, cortex, and synaptosomes isolated from these brain regions indicated changes in ephrin-related, RhoGDI, and axonal guidance signaling. Immunoblotting and miRNA-quantification demonstrated an imbalance in the synapse morphology-related Rac1-Cofilin pathway and long-term potentiation-related cAMP response element-binding protein (CREB) signaling. Proteome profiling also showed impaired oxidative phosphorylation, especially in the synaptic mitochondria. This was accompanied by an early (4 weeks) reduction of mitochondrial respiration capacity in the hippocampus. Although the respiratory capacity was restored by 24 weeks, the number of deregulated mitochondrial complex proteins was increased at this time. All observed changes were significant at doses of 0.5 and 2.0 Gy but not at 0.1 Gy. This study strongly suggests that ionizing radiation at the neonatal state triggers persistent proteomic alterations associated with synaptic impairment.
Collapse
Affiliation(s)
| | - Sara Sepe
- Department of Genetics, Erasmus Medical Center , 3015 CE Rotterdam, The Netherlands
| | | | | | | | | | - Michael J Atkinson
- Chair of Radiation Biology, Technical University Munich , 80333 Munich, Germany
| | | | | |
Collapse
|
127
|
Sandman CA. Fetal exposure to placental corticotropin-releasing hormone (pCRH) programs developmental trajectories. Peptides 2015; 72:145-53. [PMID: 25841879 PMCID: PMC4777695 DOI: 10.1016/j.peptides.2015.03.020] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 03/22/2015] [Accepted: 03/25/2015] [Indexed: 12/16/2022]
Abstract
The maternal endocrine stress system is profoundly altered during the course of human pregnancy. The human placenta expresses the genes for CRH as early as the seventh week of gestation and it is the expotential increase in placental CRH (pCRH) over the course of human gestation that is responsible for the greatest modification in the maternal stress system. The bi-directional placental release of hormones into the maternal and fetal compartments has profound influences for both. The influential Fetal Programming model predicted that early or fetal exposures to maternal signals of threat or adverse conditions have lifelong consequences for health outcomes. A basic assumption of this model was that developing organisms play a dynamic role in their own construction. Data are reviewed and new data are presented that elevated pCRH over the course of human gestation plays a fundamental role in the organization of the fetal nervous system, modifies birth phenotype (the timing of the onset of spontaneous labor and delivery), and influences developmental, temperamental and metabolic trajectories. Evidence for sex differences and conserved function across species is presented. Finally, a model is presented that proposes several pathways that pCRH can program risk for health and disease.
Collapse
Affiliation(s)
- Curt A Sandman
- Department of Psychiatry and Human Behavior, Early Human and Lifespan Development Program, University of California, Irvine, United States.
| |
Collapse
|
128
|
Bender J, Engeholm M, Ederer MS, Breu J, Møller TC, Michalakis S, Rasko T, Wanker EE, Biel M, Martinez KL, Wurst W, Deussing JM. Corticotropin-Releasing Hormone Receptor Type 1 (CRHR1) Clustering with MAGUKs Is Mediated via Its C-Terminal PDZ Binding Motif. PLoS One 2015; 10:e0136768. [PMID: 26352593 PMCID: PMC4564177 DOI: 10.1371/journal.pone.0136768] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 08/07/2015] [Indexed: 01/24/2023] Open
Abstract
The corticotropin-releasing hormone receptor type 1 (CRHR1) plays an important role in orchestrating neuroendocrine, behavioral, and autonomic responses to stress. To identify molecules capable of directly modulating CRHR1 signaling, we performed a yeast-two-hybrid screen using the C-terminal intracellular tail of the receptor as bait. We identified several members of the membrane-associated guanylate kinase (MAGUK) family: postsynaptic density protein 95 (PSD95), synapse-associated protein 97 (SAP97), SAP102 and membrane associated guanylate kinase, WW and PDZ domain containing 2 (MAGI2). CRHR1 is co-expressed with the identified MAGUKs and with the additionally investigated PSD93 in neurons of the adult mouse brain and in primary hippocampal neurons, supporting the probability of a physiological interaction in vivo. The C-terminal PDZ (PSD-95, discs large, zona occludens 1) binding motif of CRHR1 is essential for its physical interaction with MAGUKs, as revealed by the CRHR1-STAVA mutant, which harbors a functionally impaired PDZ binding motif. The imitation of a phosphorylation at Thr413 within the PDZ binding motif also disrupted the interaction with MAGUKs. In contrast, distinct PDZ domains within the identified MAGUKs are involved in the interactions. Expression of CRHR1 in primary neurons demonstrated its localization throughout the neuronal plasma membrane, including the excitatory post synapse, where the receptor co-localized with PSD95 and SAP97. The co-expression of CRHR1 and respective interacting MAGUKs in HEK293 cells resulted in a clustered subcellular co-localization which required an intact PDZ binding motif. In conclusion, our study characterized the PDZ binding motif-mediated interaction of CRHR1 with multiple MAGUKs, which directly affects receptor function.
Collapse
Affiliation(s)
- Julia Bender
- Max Planck Institute of Psychiatry, Department of Stress Neurobiology and Neurogenetics, Molecular Neurogenetics, Munich, Germany
| | | | - Marion S. Ederer
- Max Planck Institute of Psychiatry, Department of Stress Neurobiology and Neurogenetics, Molecular Neurogenetics, Munich, Germany
| | | | - Thor C. Møller
- University of Copenhagen, Department of Chemistry & Nano-Science Center, Copenhagen, Denmark
| | - Stylianos Michalakis
- Center for Integrated Protein Science Munich (CIPSM) and Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Tamas Rasko
- Max Delbrueck Center for Molecular Medicine, Berlin-Buch, Germany
| | - Erich E. Wanker
- Max Delbrueck Center for Molecular Medicine, Berlin-Buch, Germany
| | - Martin Biel
- Center for Integrated Protein Science Munich (CIPSM) and Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Karen L. Martinez
- University of Copenhagen, Department of Chemistry & Nano-Science Center, Copenhagen, Denmark
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Neurodegenerative Diseases within the Helmholtz Association, Munich, Germany
- Technische Universität München-Weihenstephan, Lehrstuhl für Entwicklungsgenetik c/o Helmholtz Zentrum München, Neuherberg, Germany
| | - Jan M. Deussing
- Max Planck Institute of Psychiatry, Department of Stress Neurobiology and Neurogenetics, Molecular Neurogenetics, Munich, Germany
- * E-mail:
| |
Collapse
|
129
|
Xu Y, Cheng X, Cui X, Wang T, Liu G, Yang R, Wang J, Bo X, Wang S, Zhou W, Zhang Y. Effects of 5-h multimodal stress on the molecules and pathways involved in dendritic morphology and cognitive function. Neurobiol Learn Mem 2015; 123:225-38. [DOI: 10.1016/j.nlm.2015.06.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 06/17/2015] [Accepted: 06/23/2015] [Indexed: 11/25/2022]
|
130
|
Busceti CL, Pietro PD, Riozzi B, Traficante A, Biagioni F, Nisticò R, Fornai F, Battaglia G, Nicoletti F, Bruno V. 5-HT2C serotonin receptor blockade prevents tau protein hyperphosphorylation and corrects the defect in hippocampal synaptic plasticity caused by a combination of environmental stressors in mice. Pharmacol Res 2015; 99:258-68. [DOI: 10.1016/j.phrs.2015.06.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 06/26/2015] [Accepted: 06/28/2015] [Indexed: 10/23/2022]
|
131
|
Sardari M, Rezayof A, Khodagholi F. Hippocampal signaling pathways are involved in stress-induced impairment of memory formation in rats. Brain Res 2015; 1625:54-63. [PMID: 26301822 DOI: 10.1016/j.brainres.2015.08.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/11/2015] [Accepted: 08/16/2015] [Indexed: 01/28/2023]
Abstract
Stress is a potent modulator of hippocampal-dependent memory formation. The aim of the present study was to assess the role of hippocampal signaling pathways in stress-induced memory impairment in male Wistar rats. The animals were exposed to acute elevated platform (EP) stress and memory formation was measured by a step-through type passive avoidance task. The results indicated that post-training or pre-test exposure to EP stress impaired memory consolidation or retrieval respectively. Using western blot analysis, it was found that memory retrieval was associated with the increase in the levels of phosphorylated cAMP-responsive element binding protein (P-CREB), peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) and its downstream targets in the hippocampus. In contrast, the stress exposure decreased the hippocampal levels of these proteins. In addition, stress-induced impairment of memory consolidation or retrieval was associated with the decrease in the P-CREB/CREB ratio and the PGC-1α level in the hippocampus. On the other hand, the hippocampal level of nuclear factor E2-related factor 2 (Nrf2) and gamma-glutamylcysteine synthetase (γ-GCS) which are the master regulators of defense system were decreased by the stress exposure. The increased hippocampal levels of Nrf2 and it׳s downstream was observed during memory retrieval, while stress-induced impairment of memory consolidation or retrieval inhibited this hippocampal signaling pathway. Overall, these findings suggest that down-regulation of CREB/PGC-1α signaling cascade and Nrf2 antioxidant pathways in the hippocampus may be associated with memory impairment induced by stress.
Collapse
Affiliation(s)
- Maryam Sardari
- Department of Animal Biology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran
| | - Ameneh Rezayof
- Department of Animal Biology, School of Biology and Center of Excellence in Phylogeny of Living Organisms, College of Science, University of Tehran, Tehran, Iran.
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
132
|
Hooper A, Maguire J. Characterization of a novel subtype of hippocampal interneurons that express corticotropin-releasing hormone. Hippocampus 2015; 26:41-53. [PMID: 26135556 DOI: 10.1002/hipo.22487] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/24/2015] [Indexed: 12/23/2022]
Abstract
A subset of corticotropin-releasing hormone (CRH) neurons was previously identified in the hippocampus with unknown function. Here we demonstrate that hippocampal CRH neurons represent a novel subtype of interneurons in the hippocampus, exhibiting unique morphology, electrophysiological properties, molecular markers, and connectivity. This subset of hippocampal CRH neurons in the mouse reside in the CA1 pyramidal cell layer and tract tracing studies using AAV-Flex-ChR2-tdTomato reveal dense back-projections of these neurons onto principal neurons in the CA3 region of the hippocampus. These hippocampal CRH neurons express both GABA and GAD67 and using in vitro optogenetic techniques, we demonstrate that these neurons make functional connections and release GABA onto CA3 principal neurons. The location, morphology, and importantly the functional connectivity of these neurons demonstrate that hippocampal CRH neurons represent a unique subtype of hippocampal interneurons. The connectivity of these neurons has significant implications for hippocampal function.
Collapse
Affiliation(s)
- Andrew Hooper
- Graduate Program in Neuroscience, Department of Neuroscience, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts
| | - Jamie Maguire
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts
| |
Collapse
|
133
|
Baglietto-Vargas D, Chen Y, Suh D, Ager RR, Rodriguez-Ortiz CJ, Medeiros R, Myczek K, Green KN, Baram TZ, LaFerla FM. Short-term modern life-like stress exacerbates Aβ-pathology and synapse loss in 3xTg-AD mice. J Neurochem 2015; 134:915-26. [PMID: 26077803 DOI: 10.1111/jnc.13195] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Revised: 05/16/2015] [Accepted: 05/22/2015] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurological disorder that impairs memory and other cognitive functions in the elderly. The social and financial impacts of AD are overwhelming and are escalating exponentially as a result of population aging. Therefore, identifying AD-related risk factors and the development of more efficacious therapeutic approaches are critical to cure this neurological disorder. Current epidemiological evidence indicates that life experiences, including chronic stress, are a risk for AD. However, it is unknown if short-term stress, lasting for hours, influences the onset or progression of AD. Here, we determined the effect of short-term, multi-modal 'modern life-like' stress on AD pathogenesis and synaptic plasticity in mice bearing three AD mutations (the 3xTg-AD mouse model). We found that combined emotional and physical stress lasting 5 h severely impaired memory in wild-type mice and tended to impact it in already low-performing 3xTg-AD mice. This stress reduced the number of synapse-bearing dendritic spines in 3xTg-AD mice and increased Aβ levels by augmenting AβPP processing. Thus, short-term stress simulating modern-life conditions may exacerbate cognitive deficits in preclinical AD by accelerating amyloid pathology and reducing synapse numbers. Epidemiological evidence indicates that life experiences, including chronic stress, are a risk for Alzheimer disease (AD). However, it is unknown if short stress in the range of hours influences the onset or progression of AD. Here, we determined the effect of short, multi-modal 'modern-lifelike'stress on AD pathogenesis and synaptic plasticity in mice bearing three AD mutations (the 3xTg-AD mouse model). We found that combined emotional and physical stress lasting 5 h severely impaired memory in wild-type mice and tended to impact it in already low-performing 3xTg-AD mice. This stress reduced the number of synapse-bearing dendritic spines in 3xTg-AD mice and increased Aβ levels by augmenting AβPP processing. Thus, short stress simulating modern-life conditions may exacerbate cognitive deficits in preclinical AD by accelerating amyloid pathology and reducing synapse numbers.
Collapse
Affiliation(s)
- David Baglietto-Vargas
- Department of Neurobiology and Behavior and Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, California, USA
| | - Yuncai Chen
- Department of Pediatrics, University of California, Irvine, California, USA
| | - Dongjin Suh
- Department of Neurobiology and Behavior and Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, California, USA
| | - Rahasson R Ager
- Department of Neurobiology and Behavior and Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, California, USA
| | - Carlos J Rodriguez-Ortiz
- Department of Neurobiology and Behavior and Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, California, USA
| | - Rodrigo Medeiros
- Department of Neurobiology and Behavior and Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, California, USA
| | - Kristoffer Myczek
- Department of Neurobiology and Behavior and Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, California, USA
| | - Kim N Green
- Department of Neurobiology and Behavior and Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, California, USA
| | - Tallie Z Baram
- Department of Pediatrics, University of California, Irvine, California, USA.,Department of Anatomy/Neurobiology, University of California, Irvine, California, USA
| | - Frank M LaFerla
- Department of Neurobiology and Behavior and Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, California, USA
| |
Collapse
|
134
|
Shu YM, Ni RJ, Sun YJ, Fang H, Zhou JN. Distribution of corticotropin-releasing factor in the tree shrew brain. Brain Res 2015; 1618:270-85. [PMID: 26074350 DOI: 10.1016/j.brainres.2015.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 05/19/2015] [Accepted: 06/04/2015] [Indexed: 11/16/2022]
Abstract
Corticotropin-releasing factor (CRF) in the brain plays an important role in regulations of physiological and behavioral processes, yet CRF distribution in tree shrew brain has not been thoroughly and systematically reported. Here we examined the distribution of CRF immunoreactivity in the brain of tree shrews (Tupaia belangeri chinensis) using immunohistochemical techniques. CRF-immunoreactive (-ir) cells and fibers were present in the rhinencephalon, telencephalon, diencephalon, mesencephalon, metencephalon and myelencephalon of saline- and colchicine-treated tree shrews. Laminar distribution of CRF-ir cells was found in the main olfactory bulb and neocortex. Compared with saline-treated tree shrews, a larger number of CRF-ir cells in colchicine-treated tree shrews were found in the bed nucleus of the stria terminalis, paraventricular hypothalamic nucleus, medial preoptic area, dorsomedial hypothalamic nucleus, reuniens thalamic nucleus, inferior colliculus, Edinger-Westphal nucleus, median raphe nucleus, locus coeruleus, parabrachial nucleus, dorsal tegmental nucleus, lateral reticular nucleus, and inferior olive. CRF-ir fibers from the hypothalamic paraventricular nucleus projected toward and through the internal zone of the median eminence. In addition, density of CRF immunoreactivity is significantly different in the bed nucleus of the stria terminalis, central amygdaloid nucleus, suprachiasmatic nucleus, median raphe nucleus, Edinger-Westphal nucleus, locus coeruleus and inferior olive between tree shrews and rats after saline or colchicine treatment. Our findings provide, for the first time, the comprehensive description of CRF immunoreactivity and whole brain mapping of CRF in tree shrews, which is an anatomical basis for the participation of CRF system in the regulation of numerous behaviors.
Collapse
Affiliation(s)
- Yu-Mian Shu
- Chinese Academy of Science Key Laboratory of Brain Function and Diseases, School of Life Sciences, University of Science and Technology of China, Hefei 230027, Anhui, PR China
| | - Rong-Jun Ni
- Chinese Academy of Science Key Laboratory of Brain Function and Diseases, School of Life Sciences, University of Science and Technology of China, Hefei 230027, Anhui, PR China
| | - Yun-Jun Sun
- Chinese Academy of Science Key Laboratory of Brain Function and Diseases, School of Life Sciences, University of Science and Technology of China, Hefei 230027, Anhui, PR China
| | - Hui Fang
- Chinese Academy of Science Key Laboratory of Brain Function and Diseases, School of Life Sciences, University of Science and Technology of China, Hefei 230027, Anhui, PR China
| | - Jiang-Ning Zhou
- Chinese Academy of Science Key Laboratory of Brain Function and Diseases, School of Life Sciences, University of Science and Technology of China, Hefei 230027, Anhui, PR China.
| |
Collapse
|
135
|
Brunton PJ. Programming the brain and behaviour by early-life stress: a focus on neuroactive steroids. J Neuroendocrinol 2015; 27:468-80. [PMID: 25688636 DOI: 10.1111/jne.12265] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 02/11/2015] [Accepted: 02/11/2015] [Indexed: 01/09/2023]
Abstract
Animal studies have amply demonstrated that stress exposure during pregnancy or in early postnatal life can adversely influence brain development and have long-term 'programming' effects on future brain function and behaviour. Furthermore, a growing body of evidence from human studies supports the hypothesis that some psychiatric disorders may have developmental origins. Here, the focus is on three adverse consequences of early-life stress: dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, heightened anxiety behaviour and cognitive impairments, with review of what is known about the underlying central mechanisms. Neuroactive steroids modulate neuronal activity and play a key role in neurodevelopment. Moreover they can negatively modulate activity of the HPA axis, exert anxiolytic actions and influence cognitive performance. Thus, neuroactive steroids may provide a link between early-life stress and the resultant adverse effects on the brain and behaviour. Here, a role for neuroactive steroids, in particular the 5α-reduced/3α-hydroxylated metabolites of progesterone, testosterone and deoxycorticosterone, is discussed in the context of early-life stress. Furthermore, the impact of early-life stress on the brain's capacity to generate neurosteroids is considered and the evidence for an ability of neuroactive steroids to over-write the negative effects of early-life stress on the brain and behaviour is examined. An enhanced understanding of the influence of early-life stress on brain neurosteroid systems could aid the identification of new targets for developing treatments for stress-related conditions in humans.
Collapse
Affiliation(s)
- P J Brunton
- Division of Neurobiology, The Roslin Institute & R(D)SVS, University of Edinburgh, Midlothian, UK
| |
Collapse
|
136
|
Croston R, Branch CL, Kozlovsky DY, Roth TC, LaDage LD, Freas CA, Pravosudov VV. Potential Mechanisms Driving Population Variation in Spatial Memory and the Hippocampus in Food-caching Chickadees. Integr Comp Biol 2015; 55:354-71. [DOI: 10.1093/icb/icv029] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
137
|
O'Doherty DCM, Chitty KM, Saddiqui S, Bennett MR, Lagopoulos J. A systematic review and meta-analysis of magnetic resonance imaging measurement of structural volumes in posttraumatic stress disorder. Psychiatry Res 2015; 232:1-33. [PMID: 25735885 DOI: 10.1016/j.pscychresns.2015.01.002] [Citation(s) in RCA: 232] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 11/15/2014] [Accepted: 01/08/2015] [Indexed: 12/15/2022]
Abstract
Posttraumatic stress disorder (PTSD) is a debilitating condition associated with mild to moderate cognitive impairment and with a prevalence rate of up to 22% in veterans. This systematic review and quantitative meta-analysis explore volumetric differences of three key structural brain regions (hippocampus, amygdala and anterior cingulate cortex (ACC)), all of which have been implicated in dysfunction of both salience network (SN) and default mode network (DMN) in PTSD sufferers. A literature search was conducted in Embase, Medline, PubMed and PsycINFO in May 2013. Fifty-nine volumetric analyses from 44 articles were examined and included (36 hippocampus, 14 amygdala and nine ACC) with n=846 PTSD participants, n=520 healthy controls (HCs) and n=624 traumatised controls (TCs). Nine statistical tests were performed for each of the three regions of interest (ROIs), measuring volume differences in PTSD subjects, healthy and traumatised controls. Hippocampal volume was reduced in subjects with PTSD, with a greater reduction in the left hippocampus. A medium effect size reduction was found in bilateral amygdala volume when compared with findings in healthy controls; however, no significant differences in amygdala volume between PTSD subjects and trauma-exposed controls were found. Significant volume reductions were found bilaterally in the ACC. While often well matched with their respective control groups, the samples of PTSD subjects composed from the source studies used in the meta-analyses are limited in their homogeneity. The current findings of reduced hippocampal volume in subjects with PTSD are consistent with the existing literature. Amygdala volumes did not show significant reductions in PTSD subjects when compared with volumes in trauma-exposed controls-congruous with reported symptoms of hypervigilance and increased propensity in acquisition of conditioned fear memories-but a significant reduction was found in the combined left and right hemisphere volume analysis when compared with healthy controls. Bilateral volume reductions in the ACC may underpin the attentional deficits and inabilities to modulate emotions that are characteristically associated with PTSD patients.
Collapse
Affiliation(s)
- Daniel C M O'Doherty
- Brain and Mind Research Institute, University of Sydney, 100 Mallett Street, Camperdown, NSW 2050, Australia.
| | - Kate M Chitty
- Brain and Mind Research Institute, University of Sydney, 100 Mallett Street, Camperdown, NSW 2050, Australia.
| | - Sonia Saddiqui
- Faculty of Human Sciences, Macquarie University, Sydney, Australia.
| | - Maxwell R Bennett
- Brain and Mind Research Institute, University of Sydney, 100 Mallett Street, Camperdown, NSW 2050, Australia.
| | - Jim Lagopoulos
- Brain and Mind Research Institute, University of Sydney, 100 Mallett Street, Camperdown, NSW 2050, Australia.
| |
Collapse
|
138
|
Jung G, Kim EJ, Cicvaric A, Sase S, Gröger M, Höger H, Sialana FJ, Berger J, Monje FJ, Lubec G. Drebrin depletion alters neurotransmitter receptor levels in protein complexes, dendritic spine morphogenesis and memory-related synaptic plasticity in the mouse hippocampus. J Neurochem 2015; 134:327-39. [DOI: 10.1111/jnc.13119] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/16/2015] [Accepted: 03/27/2015] [Indexed: 12/19/2022]
Affiliation(s)
- Gangsoo Jung
- Department of Pediatrics; Medical University of Vienna; Vienna Austria
| | - Eun-Jung Kim
- Department of Pediatrics; Medical University of Vienna; Vienna Austria
| | - Ana Cicvaric
- Department of Neurophysiology and Neuropharmacology; Center for Physiology and Pharmacology; Medical University of Vienna; Vienna Austria
| | - Sunetra Sase
- Department of Pediatrics; Medical University of Vienna; Vienna Austria
| | - Marion Gröger
- Core Facility Imaging; Medical University of Vienna; Vienna Austria
| | - Harald Höger
- Core Unit of Biomedical Research; Division of Laboratory Animal Science and Genetics; Medical University of Vienna; Himberg Austria
| | | | - Johannes Berger
- Department of Pathobiology of the Nervous System; Center for Brain Research; Medical University of Vienna; Vienna Austria
| | - Francisco J. Monje
- Department of Neurophysiology and Neuropharmacology; Center for Physiology and Pharmacology; Medical University of Vienna; Vienna Austria
| | - Gert Lubec
- Department of Pediatrics; Medical University of Vienna; Vienna Austria
| |
Collapse
|
139
|
Grimm S, Gärtner M, Fuge P, Fan Y, Weigand A, Feeser M, Aust S, Heekeren HR, Jacobs A, Heuser I, Bajbouj M. Variation in the corticotropin-releasing hormone receptor 1 (CRHR1) gene modulates age effects on working memory. J Psychiatr Res 2015; 61:57-63. [PMID: 25541005 DOI: 10.1016/j.jpsychires.2014.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 11/16/2014] [Accepted: 12/03/2014] [Indexed: 01/13/2023]
Abstract
Decline in working memory (WM) functions during aging has been associated with hippocampal dysfunction mediated by age-related changes to the corticotropin-releasing hormone (CRH) system. Recent reports suggest that GG-homozygous individuals of single nucleotide polymorphisms (rs110402 and rs242924) in the CRH receptor 1 (CRHR1) gene show increased stress vulnerability and decreased BOLD responses in WM relevant regions. However, until now, no study investigated the interaction effects of variation in the CRHR1 gene and age on individual differences in WM. Here, young, middle-aged and old subjects (N = 466) were genotyped for rs110402 and rs242924 within the CRHR1 gene and an n-back task was used to investigate the hypothesis that vulnerable genotypes (GG-homozygotes) would show impaired WM functions that might be magnified by increased CRH production with advancing age. Our results show an impact of genotype already in middle-age with significantly better performance in AT-carriers. Working memory performance in AT-carriers did not differ between young and middle-aged subjects, but was significantly impaired in old age. In GG-homozygotes, severe working memory dysfunction occurred already in middle age. Our data indicate that GG-homozygotes of CRHR1 rs110402 and rs242924 represent a genetically driven subtype of early WM impairments due to alterations in hippocampal CRHR1 activation. Early interventions that have proven effective in delaying cognitive decline appear to be particularly important for these subjects at risk for premature memory decline, who are in the prime of their personal and professional lives.
Collapse
Affiliation(s)
- Simone Grimm
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany; Department of Psychiatry, Psychotherapy and Psychosomatics, Hospital of Psychiatry, University of Zurich, 8032 Zurich, Switzerland.
| | - Matti Gärtner
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany; Department of Psychiatry, Campus Benjamin Franklin, Charité Berlin, 14050 Berlin, Germany
| | - Philipp Fuge
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany; Department of Psychiatry, Campus Benjamin Franklin, Charité Berlin, 14050 Berlin, Germany
| | - Yan Fan
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany
| | - Anne Weigand
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany; Department of Psychiatry, Campus Benjamin Franklin, Charité Berlin, 14050 Berlin, Germany
| | - Melanie Feeser
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany
| | - Sabine Aust
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany; Department of Psychiatry, Campus Benjamin Franklin, Charité Berlin, 14050 Berlin, Germany
| | - Hauke R Heekeren
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany
| | - Arthur Jacobs
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany
| | - Isabella Heuser
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany; Department of Psychiatry, Campus Benjamin Franklin, Charité Berlin, 14050 Berlin, Germany
| | - Malek Bajbouj
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, 14195 Berlin, Germany; Department of Psychiatry, Campus Benjamin Franklin, Charité Berlin, 14050 Berlin, Germany
| |
Collapse
|
140
|
Hall BS, Moda RN, Liston C. Glucocorticoid Mechanisms of Functional Connectivity Changes in Stress-Related Neuropsychiatric Disorders. Neurobiol Stress 2015; 1:174-183. [PMID: 25729760 PMCID: PMC4340078 DOI: 10.1016/j.ynstr.2014.10.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Stress—especially chronic, uncontrollable stress—is an important risk factor for many neuropsychiatric disorders. The underlying mechanisms are complex and multifactorial, but they involve correlated changes in structural and functional measures of neuronal connectivity within cortical microcircuits and across neuroanatomically distributed brain networks. Here, we review evidence from animal models and human neuroimaging studies implicating stress-associated changes in functional connectivity in the pathogenesis of PTSD, depression, and other neuropsychiatric conditions. Changes in fMRI measures of corticocortical connectivity across distributed networks may be caused by specific structural alterations that have been observed in the prefrontal cortex, hippocampus, and other vulnerable brain regions. These effects are mediated in part by glucocorticoids, which are released from the adrenal gland in response to a stressor and also oscillate in synchrony with diurnal rhythms. Recent work indicates that circadian glucocorticoid oscillations act to balance synapse formation and pruning after learning and during development, and chronic stress disrupts this balance. We conclude by considering how disrupted glucocorticoid oscillations may contribute to the pathophysiology of depression and PTSD in vulnerable individuals, and how circadian rhythm disturbances may affect non-psychiatric populations, including frequent travelers, shift workers, and patients undergoing treatment for autoimmune disorders.
Collapse
Affiliation(s)
- Baila S Hall
- Brain and Mind Research Institute and Program in Neuroscience, Weill Cornell Medical College, 413 East 69 Street, Box 240, New York, NY 10021
| | - Rachel N Moda
- Brain and Mind Research Institute and Program in Neuroscience, Weill Cornell Medical College, 413 East 69 Street, Box 240, New York, NY 10021
| | - Conor Liston
- Brain and Mind Research Institute and Program in Neuroscience, Weill Cornell Medical College, 413 East 69 Street, Box 240, New York, NY 10021 ; Sackler Institute for Developmental Psychobiology, Weill Cornell Medical College, 413 East 69 Street, Box 240, New York, NY 10021 ; Department of Psychiatry, Weill Cornell Medical College, 413 East 69 Street, Box 240, New York, NY 10021
| |
Collapse
|
141
|
Yam KY, Naninck EFG, Schmidt MV, Lucassen PJ, Korosi A. Early-life adversity programs emotional functions and the neuroendocrine stress system: the contribution of nutrition, metabolic hormones and epigenetic mechanisms. Stress 2015; 18:328-42. [PMID: 26260665 DOI: 10.3109/10253890.2015.1064890] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Clinical and pre-clinical studies have shown that early-life adversities, such as abuse or neglect, can increase the vulnerability to develop psychopathologies and cognitive decline later in life. Remarkably, the lasting consequences of stress during this sensitive period on the hypothalamic-pituitary-adrenal axis and emotional function closely resemble the long-term effects of early malnutrition and suggest a possible common pathway mediating these effects. During early-life, brain development is affected by both exogenous factors, like nutrition and maternal care as well as by endogenous modulators including stress hormones. These elements, while mostly considered for their independent actions, clearly do not act alone but rather in a synergistic manner. In order to better understand how the programming by early-life stress takes place, it is important to gain further insight into the exact interplay of these key elements, the possible common pathways as well as the underlying molecular mechanisms that mediate their effects. We here review evidence that exposure to both early-life stress and early-life under-/malnutrition similarly lead to life-long alterations on the neuroendocrine stress system and modify emotional functions. We further discuss how the different key elements of the early-life environment interact and affect one another and next suggest a possible role for the early-life adversity induced alterations in metabolic hormones and nutrient availability in shaping later stress responses and emotional function throughout life, possibly via epigenetic mechanisms. Such knowledge will help to develop intervention strategies, which gives the advantage of viewing the synergistic action of a more complete set of changes induced by early-life adversity.
Collapse
Affiliation(s)
- Kit-Yi Yam
- a Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam , XH Amsterdam , The Netherlands and
| | - Eva F G Naninck
- a Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam , XH Amsterdam , The Netherlands and
| | - Mathias V Schmidt
- b Department Stress Neurobiology and Neurogenetics , Max Planck Institute of Psychiatry , Munich , Germany
| | - Paul J Lucassen
- a Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam , XH Amsterdam , The Netherlands and
| | - Aniko Korosi
- a Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam , XH Amsterdam , The Netherlands and
| |
Collapse
|
142
|
Fuge P, Aust S, Fan Y, Weigand A, Gärtner M, Feeser M, Bajbouj M, Grimm S. Interaction of early life stress and corticotropin-releasing hormone receptor gene: effects on working memory. Biol Psychiatry 2014; 76:888-94. [PMID: 24931706 DOI: 10.1016/j.biopsych.2014.04.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 04/28/2014] [Accepted: 04/28/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND Early life stress (ELS) experience is associated with persisting working memory (WM) deficits; changes to the corticotropin-releasing hormone (CRH) system; and structural, functional, and epigenetic changes in the hippocampus. Single nucleotide polymorphisms in the CRH receptor 1 (CRHR1) gene interact with ELS experience to predict depression as well as neuroendocrine and neuronal reactivity. Although these findings indicate that vulnerable genotypes might also show impaired WM performance after ELS experience, no previous study investigated whether there is an interaction effect of CRHR1 polymorphisms and ELS experience on WM performance. METHODS Subjects (N = 451) were genotyped for rs110402 and rs242924 within the CRHR1 gene. We used an n-back task to investigate the hypothesis that WM performance in healthy subjects may be subtly influenced by functional differences in CRHR1 and represents an early marker of increased vulnerability after exposure to ELS. RESULTS Exposure to ELS had a particularly strong impact on WM performance in subjects with the common homozygous GG GG genotype, whereas only severe exposure to ELS interfered with WM accuracy in AT carriers. CONCLUSIONS Our data indicate that specific CRHR1 polymorphisms moderate the effect of ELS experience on WM performance. Exposure to ELS in combination with a vulnerable genotype results in subtle memory deficits in adulthood, which might develop before psychopathological symptoms.
Collapse
Affiliation(s)
- Philipp Fuge
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, Berlin, Germany; Department of Psychiatry, Campus Benjamin Franklin, Charité Berlin, Berlin, Germany
| | - Sabine Aust
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, Berlin, Germany; Department of Psychiatry, Campus Benjamin Franklin, Charité Berlin, Berlin, Germany
| | - Yan Fan
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, Berlin, Germany; Dahlem Institute for Neuroimaging of Emotion, Freie Universitaet Berlin, Berlin, Germany
| | - Anne Weigand
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, Berlin, Germany; Department of Psychiatry, Campus Benjamin Franklin, Charité Berlin, Berlin, Germany; Dahlem Institute for Neuroimaging of Emotion, Freie Universitaet Berlin, Berlin, Germany
| | - Matti Gärtner
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, Berlin, Germany; Dahlem Institute for Neuroimaging of Emotion, Freie Universitaet Berlin, Berlin, Germany
| | - Melanie Feeser
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, Berlin, Germany; Dahlem Institute for Neuroimaging of Emotion, Freie Universitaet Berlin, Berlin, Germany
| | - Malek Bajbouj
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, Berlin, Germany; Department of Psychiatry, Campus Benjamin Franklin, Charité Berlin, Berlin, Germany; Dahlem Institute for Neuroimaging of Emotion, Freie Universitaet Berlin, Berlin, Germany
| | - Simone Grimm
- Cluster of Excellence "Languages of Emotion", Freie Universitaet Berlin, Berlin, Germany; Department of Psychiatry, Campus Benjamin Franklin, Charité Berlin, Berlin, Germany; Dahlem Institute for Neuroimaging of Emotion, Freie Universitaet Berlin, Berlin, Germany; Department of Psychiatry, Psychotherapy and Psychosomatics, Hospital of Psychiatry, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
143
|
Çalışkan G, Schulz SB, Gruber D, Behr J, Heinemann U, Gerevich Z. Corticosterone and corticotropin-releasing factor acutely facilitate gamma oscillations in the hippocampus in vitro. Eur J Neurosci 2014; 41:31-44. [PMID: 25306895 DOI: 10.1111/ejn.12750] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Revised: 09/06/2014] [Accepted: 09/09/2014] [Indexed: 12/21/2022]
Abstract
Stressful experiences do not only cause peripheral changes in stress hormone levels, but also affect central structures such as the hippocampus, implicated in spatial orientation, stress evaluation, and learning and memory. It has been suggested that formation of memory traces is dependent on hippocampal gamma oscillations observed during alert behaviour and rapid eye movement sleep. Furthermore, during quiescent behaviour, sharp wave-ripple (SW-R) activity emerges. These events provide a temporal window during which reactivation of memory ensembles occur. We hypothesized that stress-responsive modulators, such as corticosterone (CORT), corticotropin-releasing factor (CRF) and the neurosteroid 3α, 21-dihydroxy-5α-pregnan-20-one (THDOC) are able to modulate gamma oscillations and SW-Rs. Using in vitro hippocampal slices, we studied acute and subacute (2 h) impact of these agents on gamma oscillations in area cornu ammonis 3 of the ventral hippocampus induced by acetylcholine (10 μm) combined with physostigmine (2 μm). CORT increased the gamma oscillations in a dose-dependent fashion. This effect was mediated by glucocorticoid receptors. Likewise, CRF augmented gamma oscillations via CRF type 1 receptor. Lastly, THDOC was found to diminish cholinergic gamma oscillations in a dose-dependent manner. Neither CORT, CRF nor THDOC modulated gamma power when pre-applied for 1 h, 2 h before the induction of gamma oscillations. Interestingly, stress-related neuromodulators had rather mild effects on spontaneous SW-R compared with their effects on gamma oscillations. These data suggest that the alteration of hippocampal gamma oscillation strength in vitro by stress-related agents is an acute process, permitting fast adaptation to new attention-requiring situations in vivo.
Collapse
Affiliation(s)
- Gürsel Çalışkan
- Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Charitéplatz 1, D-10117, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
144
|
Kyeremanteng C, MacKay JC, James JS, Kent P, Cayer C, Anisman H, Merali Z. Effects of electroconvulsive seizures on depression-related behavior, memory and neurochemical changes in Wistar and Wistar-Kyoto rats. Prog Neuropsychopharmacol Biol Psychiatry 2014; 54:170-8. [PMID: 24871798 DOI: 10.1016/j.pnpbp.2014.05.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 05/05/2014] [Accepted: 05/20/2014] [Indexed: 11/30/2022]
Abstract
BACKGROUND Investigations in healthy outbred rat strains have shown a potential role for brain-derived neurotrophic factor (BDNF) and the hypothalamic-pituitary-adrenal (HPA) axis in the antidepressant and memory side effects of electroconvulsive therapy (ECT, or ECS in animals). The Wistar-Kyoto (WKY) rat strain is used as a genetic model of depression yet no studies to date have directly compared the impact of ECS on the WKY strain to its healthy outbred control (Wistar). OBJECTIVE The objective of this study is to examine behavioral (antidepressant and retrograde memory) and neurochemical (BDNF and HPA axis) changes immediately (1day) and at a longer delay (7days) after repeated ECS (5 daily administrations) in WKY and Wistar rats. METHODS Male Wistar and WKY rats received 5days of repeated ECS or sham treatment and were assessed 1 and 7days later for 1) depression-like behavior and mobility; 2) retrograde memory; and 3) brain BDNF protein, brain corticotropin-releasing factor (CRF) and plasma corticosterone levels. RESULTS Both strains showed the expected antidepressant response and retrograde memory impairments at 1day following ECS, which were sustained at 7days. In addition, at 1day after ECS, Wistar and WKY rats showed similar elevations in brain BDNF and extra-hypothalamic CRF and no change in plasma corticosterone. At 7days after ECS, Wistar rats showed sustained elevations of brain BDNF and CRF, whereas WKY rats showed a normalization of brain BDNF, despite sustained elevations of brain CRF. CONCLUSIONS The model of 5 daily ECS was effective at eliciting behavioral and neurochemical changes in both strains. A temporal association was observed between brain CRF levels, but not BDNF, and measures of antidepressant effectiveness of ECS and retrograde memory impairments suggesting that extra-hypothalamic CRF may be a potential important contributor to these behavioral effects after repeated ECS/ECT.
Collapse
Affiliation(s)
- C Kyeremanteng
- School of Psychology, University of Ottawa, Ottawa, ON K1N 6N5, Canada; University of Ottawa Institute of Mental Health Research, Ottawa, ON K1Z 7K4, Canada
| | - J C MacKay
- School of Psychology, University of Ottawa, Ottawa, ON K1N 6N5, Canada; University of Ottawa Institute of Mental Health Research, Ottawa, ON K1Z 7K4, Canada
| | - J S James
- University of Ottawa Institute of Mental Health Research, Ottawa, ON K1Z 7K4, Canada
| | - P Kent
- University of Ottawa Institute of Mental Health Research, Ottawa, ON K1Z 7K4, Canada
| | - C Cayer
- University of Ottawa Institute of Mental Health Research, Ottawa, ON K1Z 7K4, Canada
| | - H Anisman
- Institute of Neuroscience, Carleton University, Ottawa, ON K1S 5B6, Canada; University of Ottawa Institute of Mental Health Research, Ottawa, ON K1Z 7K4, Canada
| | - Z Merali
- School of Psychology, University of Ottawa, Ottawa, ON K1N 6N5, Canada; Department of Psychiatry, University of Ottawa, Ottawa, ON K1N 6N5, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON K1N 6N5, Canada; University of Ottawa Institute of Mental Health Research, Ottawa, ON K1Z 7K4, Canada.
| |
Collapse
|
145
|
Laukova M, Alaluf LG, Serova LI, Arango V, Sabban EL. Early intervention with intranasal NPY prevents single prolonged stress-triggered impairments in hypothalamus and ventral hippocampus in male rats. Endocrinology 2014; 155:3920-33. [PMID: 25057792 DOI: 10.1210/en.2014-1192] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Intranasal administration of neuropeptide Y (NPY) is a promising treatment strategy to reduce traumatic stress-induced neuropsychiatric symptoms of posttraumatic stress disorder (PTSD). We evaluated the potential of intranasal NPY to prevent dysfunction of the hypothalamic-pituitary-adrenal (HPA) axis, a core neuroendocrine feature of PTSD. Rats were exposed to single prolonged stress (SPS), a PTSD animal model, and infused intranasally with vehicle or NPY immediately after SPS stressors. After 7 days undisturbed, hypothalamus and hippocampus, 2 structures regulating the HPA axis activity, were examined for changes in glucocorticoid receptor (GR) and CRH expression. Plasma ACTH and corticosterone, and hypothalamic CRH mRNA, were significantly higher in the vehicle but not NPY-treated group, compared with unstressed controls. Although total GR levels were not altered in hypothalamus, a significant decrease of GR phosphorylated on Ser232 and increased FK506-binding protein 5 mRNA were observed with the vehicle but not in animals infused with intranasal NPY. In contrast, in the ventral hippocampus, only vehicle-treated animals demonstrated elevated GR protein expression and increased GR phosphorylation on Ser232, specifically in the nuclear fraction. Additionally, SPS-induced increase of CRH mRNA in the ventral hippocampus was accompanied by apparent decrease of CRH peptide particularly in the CA3 subfield, both prevented by NPY. The results show that early intervention with intranasal NPY can prevent traumatic stress-triggered dysregulation of the HPA axis likely by restoring HPA axis proper negative feedback inhibition via GR. Thus, intranasal NPY has a potential as a noninvasive therapy to prevent negative effects of traumatic stress.
Collapse
Affiliation(s)
- Marcela Laukova
- Department of Biochemistry and Molecular Biology (M.L., L.G.A., L.I.S., E.L.S.), New York Medical College, Valhalla, New York 10595; and Molecular Imaging and Neuropathology Division (V.A.), New York State Psychiatric Institute, New York, New York 10032
| | | | | | | | | |
Collapse
|
146
|
Sripetchwandee J, Pipatpiboon N, Pratchayasakul W, Chattipakorn N, Chattipakorn SC. DPP-4 Inhibitor and PPARγ Agonist Restore the Loss of CA1 Dendritic Spines in Obese Insulin-resistant Rats. Arch Med Res 2014; 45:547-52. [DOI: 10.1016/j.arcmed.2014.09.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Accepted: 09/12/2014] [Indexed: 02/07/2023]
|
147
|
Preferential loss of dorsal-hippocampus synapses underlies memory impairments provoked by short, multimodal stress. Mol Psychiatry 2014; 19:811-22. [PMID: 24589888 PMCID: PMC4074447 DOI: 10.1038/mp.2014.12] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 12/21/2013] [Accepted: 01/06/2014] [Indexed: 02/06/2023]
Abstract
The cognitive effects of stress are profound, yet it is unknown if the consequences of concurrent multiple stresses on learning and memory differ from those of a single stress of equal intensity and duration. We compared the effects on hippocampus-dependent memory of concurrent, hours-long light, loud noise, jostling and restraint (multimodal stress) with those of restraint or of loud noise alone. We then examined if differences in memory impairment following these two stress types might derive from their differential impact on hippocampal synapses, distinguishing dorsal and ventral hippocampus. Mice exposed to hours-long restraint or loud noise were modestly or minimally impaired in novel object recognition, whereas similar-duration multimodal stress provoked severe deficits. Differences in memory were not explained by differences in plasma corticosterone levels or numbers of Fos-labeled neurons in stress-sensitive hypothalamic neurons. However, although synapses in hippocampal CA3 were impacted by both restraint and multimodal stress, multimodal stress alone reduced synapse numbers severely in dorsal CA1, a region crucial for hippocampus-dependent memory. Ventral CA1 synapses were not significantly affected by either stress modality. Probing the basis of the preferential loss of dorsal synapses after multimodal stress, we found differential patterns of neuronal activation by the two stress types. Cross-correlation matrices, reflecting functional connectivity among activated regions, demonstrated that multimodal stress reduced hippocampal correlations with septum and thalamus and increased correlations with amygdala and BST. Thus, despite similar effects on plasma corticosterone and on hypothalamic stress-sensitive cells, multimodal and restraint stress differ in their activation of brain networks and in their impact on hippocampal synapses. Both of these processes might contribute to amplified memory impairments following short, multimodal stress.
Collapse
|
148
|
Ranjan A, Behari J, Mallick BN. Cytomorphometric Changes in Hippocampal CA1 Neurons Exposed to Simulated Microgravity Using Rats as Model. Front Neurol 2014; 5:77. [PMID: 24904521 PMCID: PMC4032998 DOI: 10.3389/fneur.2014.00077] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 05/05/2014] [Indexed: 11/13/2022] Open
Abstract
Microgravity and sleep loss lead to cognitive and learning deficits. These behavioral alterations are likely to be associated with cytomorphological changes and loss of neurons. To understand the phenomenon, we exposed rats (225–275 g) to 14 days simulated microgravity (SMg) and compared its effects on CA1 hippocampal neuronal plasticity, with that of normal cage control rats. We observed that the mean area, perimeter, synaptic cleft, and length of active zone of CA1 hippocampal neurons significantly decreased while dendritic arborization and number of spines significantly increased in SMg group as compared with controls. The mean thickness of the postsynaptic density and total dendritic length remained unaltered. The changes may be a compensatory effect induced by exposure to microgravity; however, the effects may be transient or permanent, which need further study. These findings may be useful for designing effective prevention for those, including the astronauts, exposed to microgravity. Further, subject to confirmation, we propose that SMg exposure might be useful for recovery of stroke patients.
Collapse
Affiliation(s)
- Amit Ranjan
- School of Life Sciences, Jawaharlal Nehru University , New Delhi , India ; School of Environmental Sciences, Jawaharlal Nehru University , New Delhi , India
| | - Jitendra Behari
- School of Environmental Sciences, Jawaharlal Nehru University , New Delhi , India
| | - Birendra N Mallick
- School of Life Sciences, Jawaharlal Nehru University , New Delhi , India
| |
Collapse
|
149
|
Vallès A, Granic I, De Weerd P, Martens GJM. Molecular correlates of cortical network modulation by long-term sensory experience in the adult rat barrel cortex. Learn Mem 2014; 21:305-10. [PMID: 25171421 PMCID: PMC4024621 DOI: 10.1101/lm.034827.114] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Modulation of cortical network connectivity is crucial for an adaptive response to experience. In the rat barrel cortex, long-term sensory stimulation induces cortical network modifications and neuronal response changes of which the molecular basis is unknown. Here, we show that long-term somatosensory stimulation by enriched environment up-regulates cortical expression of neuropeptide mRNAs and down-regulates immediate-early gene (IEG) mRNAs specifically in the barrel cortex, and not in other brain regions. The present data suggest a central role of neuropeptides in the fine-tuning of sensory cortical circuits by long-term experience.
Collapse
Affiliation(s)
- Astrid Vallès
- Department of Neurocognition, Faculty of Psychology and Neuroscience, Maastricht University, 6229 EV Maastricht, The Netherlands Department of Molecular Animal Physiology, Radboud University, Donders Institute for Brain, Cognition and Behaviour (Centre for Neuroscience), Nijmegen Centre for Molecular Life Sciences, 6525 GA Nijmegen, The Netherlands
| | - Ivica Granic
- Department of Neurocognition, Faculty of Psychology and Neuroscience, Maastricht University, 6229 EV Maastricht, The Netherlands Department of Molecular Animal Physiology, Radboud University, Donders Institute for Brain, Cognition and Behaviour (Centre for Neuroscience), Nijmegen Centre for Molecular Life Sciences, 6525 GA Nijmegen, The Netherlands
| | - Peter De Weerd
- Department of Neurocognition, Faculty of Psychology and Neuroscience, Maastricht University, 6229 EV Maastricht, The Netherlands Department of Molecular Animal Physiology, Radboud University, Donders Institute for Brain, Cognition and Behaviour (Centre for Neuroscience), Nijmegen Centre for Molecular Life Sciences, 6525 GA Nijmegen, The Netherlands
| | - Gerard J M Martens
- Department of Molecular Animal Physiology, Radboud University, Donders Institute for Brain, Cognition and Behaviour (Centre for Neuroscience), Nijmegen Centre for Molecular Life Sciences, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
150
|
Regev L, Baram TZ. Corticotropin releasing factor in neuroplasticity. Front Neuroendocrinol 2014; 35:171-9. [PMID: 24145148 PMCID: PMC3965598 DOI: 10.1016/j.yfrne.2013.10.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 09/28/2013] [Accepted: 10/07/2013] [Indexed: 11/26/2022]
Abstract
Stress is among the strongest signals promoting neuroplasticity: Stress signals, indicating real or perceived danger, lead to alterations of neuronal function and often structure, designed to adapt to the changed conditions and promote survival. Corticotropin releasing factor (CRF) is expressed and released in several types of neuronal populations that are involved in cognition, emotion and the regulation of autonomic and endocrine function. CRF expressing neurons undergo functional and structural plasticity during stress and, in addition, the peptide acts via specific receptors to promote plasticity of target neurons.
Collapse
Affiliation(s)
- Limor Regev
- Departments of Anatomy/Neurobiology, University of California-Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Departments of Anatomy/Neurobiology, University of California-Irvine, Irvine, CA, USA; Department of Pediatrics, University of California-Irvine, Irvine, CA, USA.
| |
Collapse
|