1
|
Flatt EE, Alderman SL. 11β-Hydroxysteroid dehydrogenase type 2 may mediate the stress-specific effects of cortisol on brain cell proliferation in adult zebrafish (Danio rerio). J Exp Biol 2024; 227:jeb248020. [PMID: 39092490 PMCID: PMC11418181 DOI: 10.1242/jeb.248020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024]
Abstract
Stress-induced increases in cortisol can stimulate or inhibit brain cell proliferation, but the mechanisms behind these opposing effects are unknown. We tested the hypothesis that 11β-hydroxysteroid dehydrogenase type 2 (Hsd11b2), a glucocorticoid-inactivating enzyme expressed in neurogenic regions of the adult zebrafish brain, mitigates cortisol-induced changes to brain cell proliferation, using one of three stress regimes: a single 1 min air exposure (acute stress), two air exposures spaced 24 h apart (repeat acute stress) or social subordination (chronic stress). Plasma cortisol was significantly elevated 15 min after air exposure and recovered within 24 h after acute and repeat acute stress, whereas subordinate fish exhibited significant and sustained elevations relative to dominant fish for 24 h. Following acute stress, brain hsd11b2 transcript abundance was elevated up to 6 h after a single air exposure but was unchanged by repeat acute stress or social subordination. A sustained increase in brain Hsd11b2 protein levels occurred after acute stress, but not after repeat or chronic stress. Following acute and repeat acute stress, brain pcna transcript abundance (a marker of cell proliferation) exhibited a prolonged elevation, but was unaffected by social subordination. Interestingly, the number of telencephalic BrdU+ cells increased in fish after a single air exposure but was unchanged by repeat acute stress. Following acute and repeat acute stress, fish expressed lower brain glucocorticoid and mineralocorticoid receptor (gr and mr) transcript abundance while subordinate fish exhibited no changes. Taken together, these results demonstrate stressor-specific regulation of Hsd11b2 in the zebrafish brain that could modulate rates of cortisol catabolism contributing to observed differences in brain cell proliferation.
Collapse
Affiliation(s)
- E. Emma Flatt
- Department of Integrative Biology, University of Guelph, 50 Stone Road East, Guelph, ON, Canada, N1G 2W1
| | - Sarah L. Alderman
- Department of Integrative Biology, University of Guelph, 50 Stone Road East, Guelph, ON, Canada, N1G 2W1
| |
Collapse
|
2
|
Liu Y, Zhang J, Gu X, Jia S. Mapping the current trends and hotspots of adult hippocampal neurogenesis from 2004-2023: a bibliometric analysis. Front Neurosci 2024; 18:1416738. [PMID: 38957185 PMCID: PMC11217541 DOI: 10.3389/fnins.2024.1416738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 06/06/2024] [Indexed: 07/04/2024] Open
Abstract
Objective We utilized bibliometric and data visualization techniques to discern the primary research domains and emerging frontiers in the field of adult hippocampal neurogenesis (AHN). Methods We systematically searched the Web of Science database for AHN-related articles published between 2004 and 2023. The retrieved articles were filtered based on publication types (articles and reviews) and language (English). We employed CiteSpace, VOSviewer, and the online bibliometric platform (bibliometric.com) to visualize and analyze the collected data. Results In total, 1,590 AHN-related publications were discovered, exhibiting a steady increase in yearly publications over time. The United States emerged as the leading contributor in AHN research in terms of both publication quantity and national influence. Among all research institutions in the field of AHN, the University of California System exhibited the highest impact. Kempermann, Gerd was the most active author. The publications of the top three active authors primarily focused on the functions of AHN, and reversing hippocampal damage and cognitive impairment by improving AHN. An analysis of reference co-citation clustering revealed 8 distinct research clusters, and the notable ones included "adult hippocampal neurogenesis," "neurogenesis," "hippocampus," "dentate gyrus," "neural stem cell," and "depression." Additionally, a burst keyword detection indicated that 'anxiety' is a current research hotspot in the field of AHN. Conclusion This in-depth bibliographic assessment of AHN offers a deeper insight into the present research hotspots in the field. The association between AHN and cognitive diseases, such as Alzheimer's disease (AD) and anxiety, has emerged as a prominent research hotspot.
Collapse
Affiliation(s)
- Ye Liu
- The Second School of Clinical Medicine of Binzhou Medical University, Yantai, Shandong Province, China
- Department of Anesthesiology, Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Jian Zhang
- Department of Anesthesiology, The International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiyao Gu
- The Second School of Clinical Medicine of Binzhou Medical University, Yantai, Shandong Province, China
- Department of Anesthesiology, Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Shushan Jia
- The Second School of Clinical Medicine of Binzhou Medical University, Yantai, Shandong Province, China
| |
Collapse
|
3
|
Pangemanan L, Irwanto I, Maramis MM. Psychological dominant stressor modification to an animal model of depression with chronic unpredictable mild stress. Vet World 2023; 16:595-600. [PMID: 37041835 PMCID: PMC10082728 DOI: 10.14202/vetworld.2023.595-600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 02/09/2023] [Indexed: 04/13/2023] Open
Abstract
Background and Aim Chronic unpredictable mild stress (CUMS) is a protocol widely used to create an animal model of depression with food deprivation, water deprivation, and physical-dominant stressors as routine procedures. However, human depression mainly involves psychological stressors and does not always involve a lack of food and water; thus, CUMS procedures should be modified accordingly. Therefore, this study aimed to create an animal model of depression, mainly focusing on a psychologically dominant stressor without food and water deprivation. Materials and Methods The CUMS and control groups, respectively, received CUMS modification (psychologically dominant stressors without food and water deprivation) for 21 days. A 24-h sucrose preference test (SPT) was used to assess the successful creation of an animal model of depression. Daily food intake measurements, weekly weight monitoring, and weight gain calculations were performed. Either an independent sample t-test or the Mann-Whitney test was used. Results Of the 42 rats included, 39 completed the study. Chronic unpredictable mild stress procedures for 21 days significantly reduced the SPT (p < 0.05), mean body weight (p < 0.05), and weekly weight gain (p < 0.05) in the CUMS group compared to the control group. However, the weekly average food intake did not statistically differ between the two groups. Conclusion Psychological dominant CUMS modification to an animal model of depression resulted in lower SPT, body weight, and weekly weight gain in the CUMS group than in the control group.
Collapse
Affiliation(s)
- Lisa Pangemanan
- Doctoral Program of Medical Science, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Department of Child Health, Faculty of Medicine, Widya Mandala Catholic University, Surabaya, Indonesia
| | - Irwanto Irwanto
- Department of Child Health, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Corresponding author: Irwanto Irwanto, e-mail: Co-authors: LP: , MMM:
| | - Margarita M. Maramis
- Department of Psychiatry, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
4
|
Hodges TE, Lieblich SE, Rechlin RK, Galea LAM. Sex differences in inflammation in the hippocampus and amygdala across the lifespan in rats: associations with cognitive bias. Immun Ageing 2022; 19:43. [PMID: 36203171 PMCID: PMC9535862 DOI: 10.1186/s12979-022-00299-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/19/2022] [Indexed: 11/05/2022]
Abstract
Background Cognitive symptoms of major depressive disorder, such as negative cognitive bias, are more prevalent in women than in men. Cognitive bias involves pattern separation which requires hippocampal neurogenesis and is modulated by inflammation in the brain. Previously, we found sex differences in the activation of the amygdala and the hippocampus in response to negative cognitive bias in rats that varied with age. Given the association of cognitive bias to neurogenesis and inflammation, we examined associations between cognitive bias, neurogenesis in the hippocampus, and cytokine and chemokine levels in the ventral hippocampus (HPC) and basolateral amygdala (BLA) of male and female rats across the lifespan. Results After cognitive bias testing, males had more IFN-γ, IL-1β, IL-4, IL-5, and IL-10 in the ventral HPC than females in adolescence. In young adulthood, females had more IFN-γ, IL-1β, IL-6, and IL-10 in the BLA than males. Middle-aged rats had more IL-13, TNF-α, and CXCL1 in both regions than younger groups. Adolescent male rats had higher hippocampal neurogenesis than adolescent females after cognitive bias testing and young rats that underwent cognitive bias testing had higher levels of hippocampal neurogenesis than controls. Neurogenesis in the dorsal hippocampus was negatively associated with negative cognitive bias in young adult males. Conclusions Overall, the association between negative cognitive bias, hippocampal neurogenesis, and inflammation in the brain differs by age and sex. Hippocampal neurogenesis and inflammation may play greater role in the cognitive bias of young males compared to a greater role of BLA inflammation in adult females. These findings lay the groundwork for the discovery of sex-specific novel therapeutics that target region-specific inflammation in the brain and hippocampal neurogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s12979-022-00299-4. • Adolescent male rats had more hippocampal inflammation than females after cognitive bias testing. • Adult female rats had more basolateral amygdalar inflammation than males after cognitive bias testing. • HPC neurogenesis was negatively associated to cognitive bias in young adult male rats.
Collapse
Affiliation(s)
- Travis E. Hodges
- grid.17091.3e0000 0001 2288 9830Department of Psychology, University of British Columbia, Vancouver, Canada
| | - Stephanie E. Lieblich
- grid.17091.3e0000 0001 2288 9830Department of Psychology, University of British Columbia, Vancouver, Canada ,grid.17091.3e0000 0001 2288 9830Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Rebecca K. Rechlin
- grid.17091.3e0000 0001 2288 9830Department of Psychology, University of British Columbia, Vancouver, Canada ,grid.17091.3e0000 0001 2288 9830Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Liisa A. M. Galea
- grid.17091.3e0000 0001 2288 9830Department of Psychology, University of British Columbia, Vancouver, Canada ,grid.17091.3e0000 0001 2288 9830Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada ,grid.17091.3e0000 0001 2288 9830Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
| |
Collapse
|
5
|
Jones KL, Zhou M, Jhaveri DJ. Dissecting the role of adult hippocampal neurogenesis towards resilience versus susceptibility to stress-related mood disorders. NPJ SCIENCE OF LEARNING 2022; 7:16. [PMID: 35842419 PMCID: PMC9288448 DOI: 10.1038/s41539-022-00133-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 07/01/2022] [Indexed: 05/13/2023]
Abstract
Adult hippocampal neurogenesis in the developmental process of generating and integrating new neurons in the hippocampus during adulthood and is a unique form of structural plasticity with enormous potential to modulate neural circuit function and behaviour. Dysregulation of this process is strongly linked to stress-related neuropsychiatric conditions such as anxiety and depression, and efforts have focused on unravelling the contribution of adult-born neurons in regulating stress response and recovery. Chronic stress has been shown to impair this process, whereas treatment with clinical antidepressants was found to enhance the production of new neurons in the hippocampus. However, the precise role of adult hippocampal neurogenesis in mediating the behavioural response to chronic stress is not clear and whether these adult-born neurons buffer or increase susceptibility to stress-induced mood-related maladaptation remains one of the controversial issues. In this review, we appraise evidence probing the causal role of adult hippocampal neurogenesis in the regulation of emotional behaviour in rodents. We find that the relationship between adult-born hippocampal neurons and stress-related mood disorders is not linear, and that simple subtraction or addition of these neurons alone is not sufficient to lead to anxiety/depression or have antidepressant-like effects. We propose that future studies examining how stress affects unique properties of adult-born neurons, such as the excitability and the pattern of connectivity during their critical period of maturation will provide a deeper understanding of the mechanisms by which these neurons contribute to functional outcomes in stress-related mood disorders.
Collapse
Affiliation(s)
- Katherine L Jones
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
| | - Mei Zhou
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
- Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Dhanisha J Jhaveri
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia.
- Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia.
| |
Collapse
|
6
|
Neurobiological Links between Stress, Brain Injury, and Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8111022. [PMID: 35663199 PMCID: PMC9159819 DOI: 10.1155/2022/8111022] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 12/13/2022]
Abstract
Stress, which refers to a combination of physiological, neuroendocrine, behavioral, and emotional responses to novel or threatening stimuli, is essentially a defensive adaptation under physiological conditions. However, strong and long-lasting stress can lead to psychological and pathological damage. Growing evidence suggests that patients suffering from mild and moderate brain injuries and diseases often show severe neurological dysfunction and experience severe and persistent stressful events or environmental stimuli, whether in the acute, subacute, or recovery stage. Previous studies have shown that stress has a remarkable influence on key brain regions and brain diseases. The mechanisms through which stress affects the brain are diverse, including activation of endoplasmic reticulum stress (ERS), apoptosis, oxidative stress, and excitatory/inhibitory neuron imbalance, and may lead to behavioral and cognitive deficits. The impact of stress on brain diseases is complex and involves impediment of recovery, aggravation of cognitive impairment, and neurodegeneration. This review summarizes various stress models and their applications and then discusses the effects and mechanisms of stress on key brain regions—including the hippocampus, hypothalamus, amygdala, and prefrontal cortex—and in brain injuries and diseases—including Alzheimer’s disease, stroke, traumatic brain injury, and epilepsy. Lastly, this review highlights psychological interventions and potential therapeutic targets for patients with brain injuries and diseases who experience severe and persistent stressful events.
Collapse
|
7
|
Impact of stress on inhibitory neuronal circuits, our tribute to Bruce McEwen. Neurobiol Stress 2022; 19:100460. [PMID: 35734023 PMCID: PMC9207718 DOI: 10.1016/j.ynstr.2022.100460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/22/2022] [Accepted: 05/10/2022] [Indexed: 12/03/2022] Open
Abstract
This manuscript is dedicated to the memory of Bruce S. McEwen, to commemorate the impact he had on how we understand stress and neuronal plasticity, and the profound influence he exerted on our scientific careers. The focus of this review is the impact of stressors on inhibitory circuits, particularly those of the limbic system, but we also consider other regions affected by these adverse experiences. We revise the effects of acute and chronic stress during different stages of development and lifespan, taking into account the influence of the sex of the animals. We review first the influence of stress on the physiology of inhibitory neurons and on the expression of molecules related directly to GABAergic neurotransmission, and then focus on specific interneuron subpopulations, particularly on parvalbumin and somatostatin expressing cells. Then we analyze the effects of stress on molecules and structures related to the plasticity of inhibitory neurons: the polysialylated form of the neural cell adhesion molecule and perineuronal nets. Finally, we review the potential of antidepressants or environmental manipulations to revert the effects of stress on inhibitory circuits.
Collapse
|
8
|
Hodges TE, Lee GY, Noh SH, Galea LA. Sex and age differences in cognitive bias and neural activation in response to cognitive bias testing. Neurobiol Stress 2022; 18:100458. [PMID: 35586750 PMCID: PMC9109184 DOI: 10.1016/j.ynstr.2022.100458] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/28/2022] [Accepted: 05/02/2022] [Indexed: 12/29/2022] Open
Abstract
Cognitive symptoms of depression, including negative cognitive bias, are more severe in women than in men. Current treatments to reduce negative cognitive bias are not effective and sex differences in the neural activity underlying cognitive bias may play a role. Here we examined sex and age differences in cognitive bias and functional connectivity in a novel paradigm. Male and female rats underwent an 18-day cognitive bias procedure, in which they learned to discriminate between two contexts (shock paired context A, no-shock paired context B), during either adolescence (postnatal day (PD 40)), young adulthood (PD 100), or middle-age (PD 210). Cognitive bias was measured as freezing behaviour in response to an ambiguous context (context C), with freezing levels akin to the shock paired context coded as negative bias. All animals learned to discriminate between the two contexts, regardless of sex or age. However, adults (young adults, middle-aged) displayed a greater negative cognitive bias compared to adolescents, and middle-aged males had a greater negative cognitive bias than middle-aged females. Females had greater neural activation of the nucleus accumbens, amygdala, and hippocampal regions to the ambiguous context compared to males, and young rats (adolescent, young adults) had greater neural activation in these regions compared to middle-aged rats. Functional connectivity between regions involved in cognitive bias differed by age and sex, and only adult males had negative correlations between the frontal regions and hippocampal regions. These findings highlight the importance of examining age and sex when investigating the underpinnings of negative cognitive bias and lay the groundwork for determining what age- and sex-specific regions to target in future cognitive bias studies.
Collapse
Affiliation(s)
- Travis E. Hodges
- Department of Psychology, University of British Columbia, Canada
| | - Grace Y. Lee
- Department of Psychology, University of British Columbia, Canada
| | - Sophia H. Noh
- Department of Psychology, University of British Columbia, Canada
| | - Liisa A.M. Galea
- Department of Psychology, University of British Columbia, Canada
- Graduate Program in Neuroscience, University of British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| |
Collapse
|
9
|
D'Oliveira da Silva F, Azevedo Neto J, Sturaro C, Guarino A, Robert C, Gavioli EC, Calo G, Mouledous L, Ruzza C. The NOP antagonist BTRX-246040 increases stress resilience in mice without affecting adult neurogenesis in the hippocampus. Neuropharmacology 2022; 212:109077. [DOI: 10.1016/j.neuropharm.2022.109077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/04/2022] [Accepted: 04/27/2022] [Indexed: 11/24/2022]
|
10
|
Li F, Wang Y, Wang X, Zhao Y, Xie F, Qian LJ. Dynamic effects of chronic unpredictable mild stress on the hippocampal transcriptome in rats. Mol Med Rep 2022; 25:110. [PMID: 35119083 PMCID: PMC8845063 DOI: 10.3892/mmr.2022.12626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/17/2022] [Indexed: 11/06/2022] Open
Abstract
Stress causes extensive changes in hippocampal genomic expression, leading to changes in hippocampal structure and function. The dynamic changes in hippocampal gene expression caused by stress of different durations are still unknown. mRNA sequencing was used to analyze the hippocampal transcriptome of rats subjected to chronic unpredictable mild stress (CUMS) of different durations. Compared with the control, 501, 442 and 235 differentially expressed genes (DEGs) were detected in the hippocampus of rats subjected to CUMS for 3 days and 2 and 6 weeks, respectively. Gene Ontology (GO) analysis was used to determine the potential mechanism underlying the dynamic harmful effects of stress on the hippocampus; Certain GO terms of the down‑regulated DEGs in CUMS (3 days) rats were also found in the up‑regulated DEGs in CUMS (6 weeks) rats. These results showed opposing regulation patterns of DEGs between CUMS at 3 days and 6 weeks, which suggested a functional change from adaptation to damage in during the early and late stages of chronic stress. GO analysis for upregulated genes in rats subjected to CUMS for 3 days and 2 weeks suggested significant changes in 'extracellular matrix' and 'wound healing'. Upregulated genes in rats subjected to CUMS for 2 weeks were involved in changes associated with visual function. GO analysis of DEGs in rats subjected to CUMS for 6 weeks revealed increased expression of genes associated with 'apoptotic process' and 'aging' and decreased expression of those associated with inhibition of cell proliferation and cell structure. These results suggest that the early and middle stages of chronic stress primarily promote adaptive regulation and damage repair in the organism, while the late stage of chronic stress leads to damage in the hippocampus.
Collapse
Affiliation(s)
- Feng Li
- Department of Military Cognitive and Stress Medicine, Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, P.R. China
| | - Ying Wang
- Department of Military Cognitive and Stress Medicine, Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, P.R. China
| | - Xue Wang
- Department of Military Cognitive and Stress Medicine, Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, P.R. China
| | - Yun Zhao
- Department of Military Cognitive and Stress Medicine, Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, P.R. China
| | - Fang Xie
- Department of Military Cognitive and Stress Medicine, Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, P.R. China
| | - Ling-Jia Qian
- Department of Military Cognitive and Stress Medicine, Beijing Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing 100039, P.R. China
| |
Collapse
|
11
|
Hodges TE, Puri TA, Blankers SA, Qiu W, Galea LAM. Steroid hormones and hippocampal neurogenesis in the adult mammalian brain. VITAMINS AND HORMONES 2021; 118:129-170. [PMID: 35180925 DOI: 10.1016/bs.vh.2021.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Hippocampal neurogenesis persists across the lifespan in many species, including rodents and humans, and is associated with cognitive performance and the pathogenesis of neurodegenerative disease and psychiatric disorders. Neurogenesis is modulated by steroid hormones that change across development and differ between the sexes in rodents and humans. Here, we discuss the effects of stress and glucocorticoid exposure from gestation to adulthood as well as the effects of androgens and estrogens in adulthood on neurogenesis in the hippocampus. Throughout the review we highlight sex differences in the effects of steroid hormones on neurogenesis and how they may relate to hippocampal function and disease. These data highlight the importance of examining age and sex when evaluating the effects of steroid hormones on hippocampal neurogenesis.
Collapse
Affiliation(s)
- Travis E Hodges
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Tanvi A Puri
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Samantha A Blankers
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Wansu Qiu
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
12
|
Sung M, Sung SE, Kang KK, Choi JH, Lee S, Kim K, Lim JH, Lee GW, Rim HD, Kim BS, Won S, Kim K, Jang S, Seo MS, Woo J. Serum-Derived Neuronal Exosomal miRNAs as Biomarkers of Acute Severe Stress. Int J Mol Sci 2021; 22:9960. [PMID: 34576126 PMCID: PMC8470330 DOI: 10.3390/ijms22189960] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 12/11/2022] Open
Abstract
Stress is the physical and psychological tension felt by an individual while adapting to difficult situations. Stress is known to alter the expression of stress hormones and cause neuroinflammation in the brain. In this study, miRNAs in serum-derived neuronal exosomes (nEVs) were analyzed to determine whether differentially expressed miRNAs could be used as biomarkers of acute stress. Specifically, acute severe stress was induced in Sprague-Dawley rats via electric foot-shock treatment. In this acute severe-stress model, time-dependent changes in the expression levels of stress hormones and neuroinflammation-related markers were analyzed. In addition, nEVs were isolated from the serum of control mice and stressed mice at various time points to determine when brain damage was most prominent; this was found to be 7 days after foot shock. Next-generation sequencing was performed to compare neuronal exosomal miRNA at day 7 with the neuronal exosomal miRNA of the control group. From this analysis, 13 upregulated and 11 downregulated miRNAs were detected. These results show that specific miRNAs are differentially expressed in nEVs from an acute severe-stress animal model. Thus, this study provides novel insights into potential stress-related biomarkers.
Collapse
Affiliation(s)
- Minkyoung Sung
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (B.-S.K.); (S.W.); (K.K.); (S.J.)
| | - Soo-Eun Sung
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
| | - Kyung-Ku Kang
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
| | - Joo-Hee Choi
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
| | - Sijoon Lee
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
| | - KilSoo Kim
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
- Department of Veterinary Toxicology, College of Veterinary Medicine, Kyungpook National University, 80 Daehakro, Buk-gu, Daegu 41566, Korea
| | - Ju-Hyeon Lim
- New Drug Development Center, Osong Medical Innovation Foundation, Chungbuk 28160, Korea; (J.-H.L.); (G.W.L.)
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, 170 Hyonchung-ro, Namgu, Daegu 42415, Korea
| | - Gun Woo Lee
- Department of Orthopedic Surgery, Yeungnam University College of Medicine, Yeungnam University Medical Center, 170 Hyonchung-ro, Namgu, Daegu 42415, Korea
| | - Hyo-Deog Rim
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (B.-S.K.); (S.W.); (K.K.); (S.J.)
| | - Byung-Soo Kim
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (B.-S.K.); (S.W.); (K.K.); (S.J.)
| | - Seunghee Won
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (B.-S.K.); (S.W.); (K.K.); (S.J.)
| | - Kyungmin Kim
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (B.-S.K.); (S.W.); (K.K.); (S.J.)
| | - Seoyoung Jang
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (B.-S.K.); (S.W.); (K.K.); (S.J.)
| | - Min-Soo Seo
- Department of Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea; (M.S.); (S.-E.S.); (K.-K.K.); (J.-H.C.); (S.L.); (K.K.)
| | - Jungmin Woo
- Department of Psychiatry, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (H.-D.R.); (B.-S.K.); (S.W.); (K.K.); (S.J.)
| |
Collapse
|
13
|
Tzu-Feng Wang TF, Tsai SF, Zhao ZW, Shih MMC, Wang CY, Yang TT, Kuo YM. Exercise-induced increases of corticosterone contribute to exercise-enhanced adult hippocampal neurogenesis in mice. CHINESE J PHYSIOL 2021; 64:186-193. [PMID: 34472449 DOI: 10.4103/cjp.cjp_39_21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Adult hippocampal neurogenesis (AHN) is suppressed by chronic stress. The negative effect of stress is mainly attributed to increased levels of stress hormones (e.g. glucocorticoids, GCs). Exercise enhances AHN, yet it also stimulates GC secretion. To delineate the paradoxical role of GCs, we took the advantage of a unique mouse strain (L/L) which exhibits an inert response to stress-induced secretion of GCs to study the role of GCs in exercise-induced AHN. Our results showed that basal corticosterone (CORT), the main GCs in rodents, levels were similar between the L/L mice and wild-type (WT) mice. However, levels of CORT in the L/L mice were barely altered and significantly lower than those of the WT mice during treadmill running (TR). AHN was enhanced by 4 weeks of TR in the WT mice, but not L/L mice. WT mice that received daily injection of CORT to evoke serum CORT levels similar to those during exercise for 4 weeks did not affect AHN, whereas injection with large amount of CORT inhibited AHN. Taken together, our results indicated that exercise-related elevation of CORT participates in exercise-enhanced AHN. CORT alone is not sufficient to elicit AHN and may inhibit AHN if the levels are high.
Collapse
Affiliation(s)
- Tzu-Feng Tzu-Feng Wang
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei; Department of Cell Biology and Anatomy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Feng Tsai
- Department of Cell Biology and Anatomy, College of Medicine; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Zi-Wei Zhao
- Department of Cell Biology and Anatomy, College of Medicine; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | - Chia-Yih Wang
- Department of Cell Biology and Anatomy, College of Medicine; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ting-Ting Yang
- Department of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung, Taiwan
| | - Yu-Min Kuo
- Department of Cell Biology and Anatomy, College of Medicine; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
14
|
Podgorny OV, Gulyaeva NV. Glucocorticoid-mediated mechanisms of hippocampal damage: Contribution of subgranular neurogenesis. J Neurochem 2020; 157:370-392. [PMID: 33301616 DOI: 10.1111/jnc.15265] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/09/2020] [Accepted: 11/30/2020] [Indexed: 12/19/2022]
Abstract
A comprehensive overview of the interplay between glucocorticoids (GCs) and adult hippocampal neurogenesis (AHN) is presented, particularly, in the context of a diseased brain. The effectors of GCs in the dentate gyrus neurogenic niche of the hippocampal are reviewed, and the consequences of the GC signaling on the generation and integration of new neurons are discussed. Recent findings demonstrating how GC signaling mediates impairments of the AHN in various brain pathologies are overviewed. GC-mediated effects on the generation and integration of adult-born neurons in the hippocampal dentate gyrus depend on the nature, severity, and duration of the acting stress factor. GCs realize their effects on the AHN primarily via specific glucocorticoid and mineralocorticoid receptors. Disruption of the reciprocal regulation between the hypothalamic-pituitary-adrenal (HPA) axis and the generation of the adult-born granular neurons is currently considered to be a key mechanism implicating the AHN into the pathogenesis of numerous brain diseases, including those without a direct hippocampal damage. These alterations vary from reduced proliferation of stem and progenitor cells to increased cell death and abnormalities in morphology, connectivity, and localization of young neurons. Although the involvement of the mutual regulation between the HPA axis and the AHN in the pathogenesis of cognitive deficits and mood impairments is evident, several unresolved critical issues are stated. Understanding the details of GC-mediated mechanisms involved in the alterations in AHN could enable the identification of molecular targets for ameliorating pathology-induced imbalance in the HPA axis/AHN mutual regulation to conquer cognitive and psychiatric disturbances.
Collapse
Affiliation(s)
- Oleg V Podgorny
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia.,Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, Russia
| | - Natalia V Gulyaeva
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Moscow, Russia.,Research and Clinical Center for Neuropsychiatry of Moscow Healthcare Department, Moscow, Russia
| |
Collapse
|
15
|
Flores NP, Bona NP, Luduvico KP, Cardoso JDS, Soares MSP, Gamaro GD, Spanevello RM, Lencina CL, Gazal M, Stefanello FM. Eugenia uniflora fruit extract exerts neuroprotective effect on chronic unpredictable stress-induced behavioral and neurochemical changes. J Food Biochem 2020; 44:e13442. [PMID: 32803896 DOI: 10.1111/jfbc.13442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/28/2020] [Accepted: 08/03/2020] [Indexed: 01/01/2023]
Abstract
The aim of the current study was to evaluate the effect of chronic administration of Eugenia uniflora fruit extract on behavioral parameters, oxidative stress markers, and acetylcholinesterase activity in an animal model of depression, which was induced by chronic unpredictable stress (CUS). Mice were divided into six groups as follows: control/vehicle (water), control/fluoxetine (20 mg/kg), control/extract (200 mg/kg), CUS/vehicle, CUS/fluoxetine (20 mg/kg), and CUS/extract (200 mg/kg). Animals of the CUS group were exposed to a series of stressors for a period of 21 days. Vehicle, fluoxetine, and hydroalcoholic extract were administered daily by gavage. Results showed that E. uniflora treatment: (a) prevented the depressant-like effect induced by CUS; (b) regulated the activity of acetylcholinesterase; (c) reduced oxidative damage to lipids and reactive oxygen species production, in the prefrontal cortex and hippocampus; and (d) prevented the reduction of glutathione peroxidase in the hippocampus of animals subjected to CUS protocol. Taken together, our findings suggested that E. uniflora extract exerts a neuroprotective effect by preventing oxidative damage and decreasing CUS-induced acetylcholinesterase activity, thus, ameliorating depressive-type behavior. PRACTICAL APPLICATIONS: E. uniflora fruit extract revealed an antidepressant-like effect and prevented the oxidative damage as well as cholinergic alterations caused by chronic stress in mice. Therefore, we believe that the results obtained in this study can be used to develop an alternative therapy for the management of depressive disorders.
Collapse
Affiliation(s)
- Natália Porto Flores
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Natália Pontes Bona
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Karina Pereira Luduvico
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Juliane de Souza Cardoso
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Mayara Sandrielly Pereira Soares
- Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Giovana Duzzo Gamaro
- Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Roselia Maria Spanevello
- Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Claiton Leoneti Lencina
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Marta Gazal
- Programa de Biologia Celular e Molecular-Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Francieli Moro Stefanello
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Pelotas, Brazil
| |
Collapse
|
16
|
Sarkar T, Patro N, Patro IK. Neuronal changes and cognitive deficits in a multi-hit rat model following cumulative impact of early life stressors. Biol Open 2020; 9:bio054130. [PMID: 32878878 PMCID: PMC7522020 DOI: 10.1242/bio.054130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/20/2020] [Indexed: 01/06/2023] Open
Abstract
Perinatal protein malnourishment (LP) is a leading cause for mental and physical retardation in children from poor socioeconomic conditions. Such malnourished children are vulnerable to additional stressors that may synergistically act to cause neurological disorders in adulthood. In this study, the above mentioned condition was mimicked via a multi-hit rat model in which pups born to LP mothers were co-injected with polyinosinic:polycytidylic acid (Poly I:C; viral mimetic) at postnatal day (PND) 3 and lipopolysaccharide (LPS; bacterial mimetic) at PND 9. Individual exposure of Poly I:C and LPS was also given to LP pups to correlate chronicity of stress. Similar treatments were also given to control pups. Hippocampal cellular apoptosis, β III tubulin catastrophe, altered neuronal profiling and spatial memory impairments were assessed at PND 180, using specific immunohistochemical markers (active caspase 3, β III tubulin, doublecortin), golgi studies and cognitive mazes (Morris water maze and T maze). Increase in cellular apoptosis, loss of dendritic arborization and spatial memory impairments were higher in the multi-hit group, than the single-hit groups. Such impairments observed due to multi-hit stress mimicked conditions similar to many neurological disorders and hence, it is hypothesized that later life neurological disorders might be an outcome of multiple early life hits.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Tiyasha Sarkar
- School of Studies in Neuroscience, Jiwaji University, Gwalior 474011, India
| | - Nisha Patro
- School of Studies in Neuroscience, Jiwaji University, Gwalior 474011, India
| | - Ishan Kumar Patro
- School of Studies in Neuroscience, Jiwaji University, Gwalior 474011, India
| |
Collapse
|
17
|
Jorgensen C, Wang Z. Hormonal Regulation of Mammalian Adult Neurogenesis: A Multifaceted Mechanism. Biomolecules 2020; 10:biom10081151. [PMID: 32781670 PMCID: PMC7465680 DOI: 10.3390/biom10081151] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/27/2020] [Accepted: 08/01/2020] [Indexed: 02/07/2023] Open
Abstract
Adult neurogenesis—resulting in adult-generated functioning, integrated neurons—is still one of the most captivating research areas of neuroplasticity. The addition of new neurons in adulthood follows a seemingly consistent multi-step process. These neurogenic stages include proliferation, differentiation, migration, maturation/survival, and integration of new neurons into the existing neuronal network. Most studies assessing the impact of exogenous (e.g., restraint stress) or endogenous (e.g., neurotrophins) factors on adult neurogenesis have focused on proliferation, survival, and neuronal differentiation. This review will discuss the multifaceted impact of hormones on these various stages of adult neurogenesis. Specifically, we will review the evidence for hormonal facilitation (via gonadal hormones), inhibition (via glucocorticoids), and neuroprotection (via recruitment of other neurochemicals such as neurotrophin and neuromodulators) on newly adult-generated neurons in the mammalian brain.
Collapse
Affiliation(s)
- Claudia Jorgensen
- Behavioral Science Department, Utah Valley University, Orem, UT 84058, USA
- Correspondence:
| | - Zuoxin Wang
- Psychology Department and Program in Neuroscience, Florida State University, Tallahassee, FL 32306, USA;
| |
Collapse
|
18
|
Abstract
In the adult mammalian hippocampus, new neurons arise from stem and progenitor cell division, in a process known as adult neurogenesis. Adult-generated neurons are sensitive to experience and may participate in hippocampal functions, including learning and memory, anxiety and stress regulation, and social behavior. Increasing evidence emphasizes the importance of new neuron connectivity within hippocampal circuitry for understanding the impact of adult neurogenesis on brain function. In this Review, we discuss how the functional consequences of new neurons arise from the collective interactions of presynaptic and postsynaptic neurons, glial cells, and the extracellular matrix, which together form the "tetrapartite synapse."
Collapse
Affiliation(s)
- Elise C Cope
- Princeton Neuroscience Institute and Department of Psychology, Princeton University, Princeton, NJ 08544, USA
| | - Elizabeth Gould
- Princeton Neuroscience Institute and Department of Psychology, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
19
|
Abstract
Understanding the neurobiological basis of post-traumatic stress disorder (PTSD) is fundamental to accurately diagnose this neuropathology and offer appropriate treatment options to patients. The lack of pharmacological effects, too often observed with the most currently used drugs, the selective serotonin reuptake inhibitors (SSRIs), makes even more urgent the discovery of new pharmacological approaches. Reliable animal models of PTSD are difficult to establish because of the present limited understanding of the PTSD heterogeneity and of the influence of various environmental factors that trigger the disorder in humans. We summarize knowledge on the most frequently investigated animal models of PTSD, focusing on both their behavioral and neurobiological features. Most of them can reproduce not only behavioral endophenotypes, including anxiety-like behaviors or fear-related avoidance, but also neurobiological alterations, such as glucocorticoid receptor hypersensitivity or amygdala hyperactivity. Among the various models analyzed, we focus on the social isolation mouse model, which reproduces some deficits observed in humans with PTSD, such as abnormal neurosteroid biosynthesis, changes in GABAA receptor subunit expression and lack of pharmacological response to benzodiazepines. Neurosteroid biosynthesis and its interaction with the endocannabinoid system are altered in PTSD and are promising neuronal targets to discover novel PTSD agents. In this regard, we discuss pharmacological interventions and we highlight exciting new developments in the fields of research for novel reliable PTSD biomarkers that may enable precise diagnosis of the disorder and more successful pharmacological treatments for PTSD patients.
Collapse
|
20
|
Hormozi A, Zarifkar A, Rostami B, Naghibalhossaini F. An Experimental Study on Spinal Cord µ-Opioid and α2-Adrenergic Receptors mRNA Expression Following Stress-Induced Hyperalgesia in Male Rats. IRANIAN JOURNAL OF MEDICAL SCIENCES 2019; 44:397-405. [PMID: 31582864 PMCID: PMC6754534 DOI: 10.30476/ijms.2019.44958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background: Intense stress can change pain perception and induce hyperalgesia; a phenomenon called stress-induced hyperalgesia (SIH). However, the neurobiological mechanism of this effect remains unclear. The present study aimed to investigate the effect of the spinal cord µ-opioid receptors (MOR) and α2-adrenergic receptors (α2-AR) on pain sensation in rats with SIH.
Methods: Eighteen Sprague-Dawley male rats, weighing 200-250 g, were randomly divided into two groups (n=9 per group), namely the control and stress group. The stress group was evoked by random 1-hour daily foot-shock stress (0.8 mA for 10 seconds, 1 minute apart) for 3 weeks using a communication box. The tail-flick and formalin tests were performed in both groups on day 22. The real-time RT-PCR technique was used to observe MOR and α2-AR mRNA levels at the L4-L5 lumbar spinal cord. Statistical analysis was performed using the GraphPad Prism 5 software (San Diego, CA, USA). Student’s t test was applied for comparisons between the groups. P<0.05 was considered statistically significant.
Results: There was a significant (P=0.0014) decrease in tail-flick latency in the stress group compared to the control group. Nociceptive behavioral responses to formalin-induced pain in the stress group were significantly increased in the acute (P=0.007) and chronic (P=0.001) phases of the formalin test compared to the control group. A significant reduction was also observed in MOR mRNA level of the stress group compared to the control group (P=0.003). There was no significant difference in α2-AR mRNA level between the stress and control group.
Conclusion: The results indicate that chronic stress can affect nociception and lead to hyperalgesia. The data suggest that decreased expression of spinal cord MOR causes hyperalgesia.
Collapse
Affiliation(s)
- Asef Hormozi
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asadollah Zarifkar
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Neuroscience, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahar Rostami
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fakhraddin Naghibalhossaini
- Department of Biochemistry, Shiraz University of Medical Sciences, School of Medicine, Shiraz, Iran.,Autoimmune Research Center, Shiraz University of Medical Sciences, School of Medicine, Shiraz, Iran
| |
Collapse
|
21
|
The Impact of Ethologically Relevant Stressors on Adult Mammalian Neurogenesis. Brain Sci 2019; 9:brainsci9070158. [PMID: 31277460 PMCID: PMC6680763 DOI: 10.3390/brainsci9070158] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 06/30/2019] [Accepted: 07/02/2019] [Indexed: 12/18/2022] Open
Abstract
Adult neurogenesis—the formation and functional integration of adult-generated neurons—remains a hot neuroscience topic. Decades of research have identified numerous endogenous (such as neurotransmitters and hormones) and exogenous (such as environmental enrichment and exercise) factors that regulate the various neurogenic stages. Stress, an exogenous factor, has received a lot of attention. Despite the large number of reviews discussing the impact of stress on adult neurogenesis, no systematic review on ethologically relevant stressors exists to date. The current review details the effects of conspecifically-induced psychosocial stress (specifically looking at the lack or disruption of social interactions and confrontation) as well as non-conspecifically-induced stress on mammalian adult neurogenesis. The underlying mechanisms, as well as the possible functional role of the altered neurogenesis level, are also discussed. The reviewed data suggest that ethologically relevant stressors reduce adult neurogenesis.
Collapse
|
22
|
Schoenfeld TJ, Rhee D, Martin L, Smith JA, Sonti AN, Padmanaban V, Cameron HA. New neurons restore structural and behavioral abnormalities in a rat model of PTSD. Hippocampus 2019; 29:848-861. [PMID: 30865372 DOI: 10.1002/hipo.23087] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 01/24/2019] [Accepted: 02/20/2019] [Indexed: 12/19/2022]
Abstract
Post-traumatic stress disorder (PTSD) has been associated with anxiety, memory impairments, enhanced fear, and hippocampal volume loss, although the relationship between these changes remain unknown. Single-prolonged stress (SPS) is a model for PTSD combining three forms of stress (restraint, swim, and anesthesia) in a single session that results in prolonged behavioral effects. Using pharmacogenetic ablation of adult neurogenesis in rats, we investigated the role of new neurons in the hippocampus in the long-lasting structural and behavioral effects of SPS. Two weeks after SPS, stressed rats displayed increased anxiety-like behavior and decreased preference for objects in novel locations regardless of the presence or absence of new neurons. Chronic stress produced by daily restraint for 2 or 6 hr produced similar behavioral effects that were also independent of ongoing neurogenesis. At a longer recovery time point, 1 month after SPS, rats with intact neurogenesis had normalized, showing control levels of anxiety-like behavior. However, GFAP-TK rats, which lacked new neurons, continued to show elevated anxiety-like behavior and enhanced serum corticosterone response to anxiogenic experience. Volume loss in ventral CA1 region of the hippocampus paralleled increases in anxiety-like behavior, occurring in all rats exposed to SPS at the early time point and only rats lacking adult neurogenesis at the later time point. In chronic stress experiments, volume loss occurred broadly throughout the dentate gyrus and CA1 after 6-hr daily stress but was not apparent in any hippocampal subregion after 2-hr daily stress. No effect of SPS was seen on cell proliferation in the dentate gyrus, but the survival of young neurons born a week after stress was decreased. Together, these data suggest that new neurons are important for recovery of normal behavior and hippocampal structure following a strong acute stress and point to the ventral CA1 region as a potential key mediator of stress-induced anxiety-like behavior.
Collapse
Affiliation(s)
- Timothy J Schoenfeld
- Section on Neuroplasticity, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Diane Rhee
- Section on Neuroplasticity, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Laura Martin
- Section on Neuroplasticity, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Jesse A Smith
- Section on Neuroplasticity, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Anup N Sonti
- Section on Neuroplasticity, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Varun Padmanaban
- Section on Neuroplasticity, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| | - Heather A Cameron
- Section on Neuroplasticity, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
23
|
Baptista P, Andrade JP. Adult Hippocampal Neurogenesis: Regulation and Possible Functional and Clinical Correlates. Front Neuroanat 2018; 12:44. [PMID: 29922131 PMCID: PMC5996050 DOI: 10.3389/fnana.2018.00044] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 05/11/2018] [Indexed: 01/19/2023] Open
Abstract
The formation of new neurons in the adult central nervous system (CNS) has been recognized as one of the major findings in neuroanatomical research. The hippocampal formation (HF), one of the main targets of these investigations, holds a neurogenic niche widely recognized among several mammalian species and whose existence in the human brain has sparked controversy and extensive debate. Many cellular features from this region emphasize that hippocampal neurogenesis suffers changes with normal aging and, among regulatory factors, physical exercise and chronic stress provoke opposite effects on cell proliferation, maturation and survival. Considering the numerous functions attributable to the HF, increasing or decreasing the integration of new neurons in the delicate neuronal network might be significant for modulation of cognition and emotion. The role that immature and mature adult-born neurons play in this circuitry is still mostly unknown but it could prove fundamental to understand hippocampal-dependent cognitive processes, the pathophysiology of depression, and the therapeutic effects of antidepressant medication in modulating behavior and mental health.
Collapse
Affiliation(s)
- Pedro Baptista
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine of University of Porto, Porto, Portugal
| | - José P Andrade
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine of University of Porto, Porto, Portugal.,Center of Health Technology and Services Research (CINTESIS), Faculty of Medicine of University of Porto, Porto, Portugal
| |
Collapse
|
24
|
Steenbergen PJ. Response of zebrafish larvae to mild electrical stimuli: A 96-well setup for behavioural screening. J Neurosci Methods 2018. [DOI: 10.1016/j.jneumeth.2018.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
25
|
Cameron HA, Schoenfeld TJ. Behavioral and structural adaptations to stress. Front Neuroendocrinol 2018; 49:106-113. [PMID: 29421158 PMCID: PMC5963997 DOI: 10.1016/j.yfrne.2018.02.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/20/2018] [Accepted: 02/03/2018] [Indexed: 10/18/2022]
Abstract
Unpredictable aversive experiences, or stressors, lead to changes in depression- and anxiety-related behavior and to changes in hippocampal structure including decreases in adult neurogenesis, granule cell and pyramidal cell dendritic morphology, and volume. Here we review the relationship between these behavioral and structural changes and discuss the possibility that these changes may be largely adaptive. Specifically, we suggest that new neurons in the dentate gyrus enhance behavioral adaptability to changes in the environment, biasing behavior in novel situations based on previous experience with stress. Conversely, atrophy-like changes in the hippocampus and decreased adult neurogenesis following chronic stress may serve to limit stress responses and stabilize behavior during chronic stress.
Collapse
Affiliation(s)
- Heather A Cameron
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Timothy J Schoenfeld
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
26
|
Luarte A, Cisternas P, Caviedes A, Batiz LF, Lafourcade C, Wyneken U, Henzi R. Astrocytes at the Hub of the Stress Response: Potential Modulation of Neurogenesis by miRNAs in Astrocyte-Derived Exosomes. Stem Cells Int 2017; 2017:1719050. [PMID: 29081809 PMCID: PMC5610870 DOI: 10.1155/2017/1719050] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 08/16/2017] [Indexed: 01/24/2023] Open
Abstract
Repetitive stress negatively affects several brain functions and neuronal networks. Moreover, adult neurogenesis is consistently impaired in chronic stress models and in associated human diseases such as unipolar depression and bipolar disorder, while it is restored by effective antidepressant treatments. The adult neurogenic niche contains neural progenitor cells in addition to amplifying progenitors, neuroblasts, immature and mature neurons, pericytes, astrocytes, and microglial cells. Because of their particular and crucial position, with their end feet enwrapping endothelial cells and their close communication with the cells of the niche, astrocytes might constitute a nodal point to bridge or transduce systemic stress signals from peripheral blood, such as glucocorticoids, to the cells involved in the neurogenic process. It has been proposed that communication between astrocytes and niche cells depends on direct cell-cell contacts and soluble mediators. In addition, new evidence suggests that this communication might be mediated by extracellular vesicles such as exosomes, and in particular, by their miRNA cargo. Here, we address some of the latest findings regarding the impact of stress in the biology of the neurogenic niche, and postulate how astrocytic exosomes (and miRNAs) may play a fundamental role in such phenomenon.
Collapse
Affiliation(s)
- Alejandro Luarte
- Centro de Investigaciones Biomédicas, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Biomedical Neuroscience Institute, Universidad de Chile, Santiago, Chile
| | - Pablo Cisternas
- Centro de Investigaciones Biomédicas, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Cells for Cells, Santiago, Chile
| | - Ariel Caviedes
- Centro de Investigaciones Biomédicas, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Luis Federico Batiz
- Centro de Investigaciones Biomédicas, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Carlos Lafourcade
- Centro de Investigaciones Biomédicas, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Ursula Wyneken
- Centro de Investigaciones Biomédicas, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Roberto Henzi
- Centro de Investigaciones Biomédicas, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| |
Collapse
|
27
|
Lau C, Hebert M, Vani MA, Walling S, Hayley S, Lagace DC, Blundell J. Absence of neurogenic response following robust predator-induced stress response. Neuroscience 2016; 339:276-286. [DOI: 10.1016/j.neuroscience.2016.10.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 09/23/2016] [Accepted: 10/02/2016] [Indexed: 12/27/2022]
|
28
|
|
29
|
Fitzsimons CP, Herbert J, Schouten M, Meijer OC, Lucassen PJ, Lightman S. Circadian and ultradian glucocorticoid rhythmicity: Implications for the effects of glucocorticoids on neural stem cells and adult hippocampal neurogenesis. Front Neuroendocrinol 2016; 41:44-58. [PMID: 27234350 DOI: 10.1016/j.yfrne.2016.05.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 05/01/2016] [Accepted: 05/03/2016] [Indexed: 01/01/2023]
Abstract
Psychosocial stress, and within the neuroendocrine reaction to stress specifically the glucocorticoid hormones, are well-characterized inhibitors of neural stem/progenitor cell proliferation in the adult hippocampus, resulting in a marked reduction in the production of new neurons in this brain area relevant for learning and memory. However, the mechanisms by which stress, and particularly glucocorticoids, inhibit neural stem/progenitor cell proliferation remain unclear and under debate. Here we review the literature on the topic and discuss the evidence for direct and indirect effects of glucocorticoids on neural stem/progenitor cell proliferation and adult neurogenesis. Further, we discuss the hypothesis that glucocorticoid rhythmicity and oscillations originating from the activity of the hypothalamus-pituitary-adrenal axis, may be crucial for the regulation of neural stem/progenitor cells in the hippocampus, as well as the implications of this hypothesis for pathophysiological conditions in which glucocorticoid oscillations are affected.
Collapse
Affiliation(s)
- Carlos P Fitzsimons
- Neuroscience Program, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098XH Amsterdam, The Netherlands.
| | - Joe Herbert
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, United Kingdom
| | - Marijn Schouten
- Neuroscience Program, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098XH Amsterdam, The Netherlands
| | - Onno C Meijer
- Leiden University Medical Centre, Department of Endocrinology, Leiden, The Netherlands
| | - Paul J Lucassen
- Neuroscience Program, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098XH Amsterdam, The Netherlands.
| | - Stafford Lightman
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, Faculty of Medicine and Dentistry, University of Bristol, United Kingdom
| |
Collapse
|
30
|
Hanif AM, Lawson EC, Prunty M, Gogniat M, Aung MH, Chakraborty R, Boatright JH, Pardue MT. Neuroprotective Effects of Voluntary Exercise in an Inherited Retinal Degeneration Mouse Model. Invest Ophthalmol Vis Sci 2016; 56:6839-46. [PMID: 26567796 DOI: 10.1167/iovs.15-16792] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Our previous investigations showed that involuntary treadmill exercise is neuroprotective in a light-induced retinal degeneration mouse model, and it may act through activation of tropomyosin-related kinase B (TrkB) receptors. This study investigated whether voluntary running wheel exercise can be neuroprotective in an inheritable model of the retinal degenerative disease retinitis pigmentosa (RP), rd10 mice. METHODS Breeding pairs of rd10 and C57BL/6J mice were given free-spinning (active) or locked (inactive) running wheels. Pups were weaned into separate cages with their parents' respective wheel types, and visual function was tested with ERG and a virtual optokinetic system at 4, 5, and 6 weeks of age. Offspring were killed at 6 weeks of age and retinal cross-sections were prepared for photoreceptor nuclei counting. Additionally, separate cohorts of active and inactive rd10 pups were injected daily for 14 days after eye opening with a selective TrkB receptor antagonist (ANA-12) or vehicle solution and assessed as described above. RESULTS Mice in the rd10 active group exhibited significant preservation of visual acuity, cone nuclei, and total photoreceptor nuclei number. Injection with ANA-12 precluded the preservation of visual acuity and photoreceptor nuclei number in rd10 mice. CONCLUSIONS Voluntary running partially protected against the retinal degeneration and vision loss that otherwise occurs in the rd10 mouse model of RP. This protection was prevented by injection of ANA-12, suggesting that TrkB activation mediates exercise's preservation of the retina. Exercise may serve as an effective, clinically translational intervention against retinal degeneration.
Collapse
Affiliation(s)
- Adam M Hanif
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, Georgia, United States
| | - Eric C Lawson
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, Georgia, United States
| | - Megan Prunty
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, Georgia, United States
| | - Marissa Gogniat
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, Georgia, United States
| | - Moe H Aung
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Ranjay Chakraborty
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Jeffrey H Boatright
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, Georgia, United States 2Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Machelle T Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, Georgia, United States 2Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, United States
| |
Collapse
|
31
|
Distler MJ, Jungblut LD, Ceballos NR, Paz DA, Pozzi AG. Overcrowding-mediated stress alters cell proliferation in key neuroendocrine areas during larval development in Rhinella arenarum. ACTA ACUST UNITED AC 2016; 325:149-57. [PMID: 26817921 DOI: 10.1002/jez.2005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 01/11/2016] [Accepted: 01/11/2016] [Indexed: 11/07/2022]
Abstract
Exposure to adverse environmental conditions can elicit a stress response, which results in an increase in endogenous corticosterone levels. In early life stages, it has been thoroughly demonstrated that amphibian larval growth and development is altered as a consequence of chronic stress by interfering with the metamorphic process, however, the underlying mechanisms involved have only been partially disentangled. We examined the effect of intraspecific competition on corticosterone levels during larval development of the toad Rhinella arenarum and its ultimate effects on cell proliferation in particular brain areas as well as the pituitary gland. While overcrowding altered the number of proliferating cells in the pituitary gland, hypothalamus, and third ventricle of the brain, no differences were observed in areas which are less associated with neuroendocrine processes, such as the first ventricle of the brain. Apoptosis was increased in hypothalamic regions but not in the pituitary. With regards to pituitary cell populations, thyrotrophs but not somatoatrophs and corticotrophs showed a decrease in the cell number in overcrowded larvae. Our study shows that alterations in growth and development, produced by stress, results from an imbalance in the neuroendocrine systems implicated in orchestrating the timing of metamorphosis.
Collapse
Affiliation(s)
- Mijal J Distler
- Facultad de Ciencias Exactas y Naturales, Departamento de Biodiversidad y Biología Experimental, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Lucas D Jungblut
- Facultad de Ciencias Exactas y Naturales, Departamento de Biodiversidad y Biología Experimental, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Fisiología, Biología Molecular y Neurociencias, CONICET, Buenos Aires, Argentina
| | - Nora R Ceballos
- Facultad de Ciencias Exactas y Naturales, Departamento de Biodiversidad y Biología Experimental, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Dante A Paz
- Facultad de Ciencias Exactas y Naturales, Departamento de Biodiversidad y Biología Experimental, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Fisiología, Biología Molecular y Neurociencias, CONICET, Buenos Aires, Argentina
| | - Andrea G Pozzi
- Facultad de Ciencias Exactas y Naturales, Departamento de Biodiversidad y Biología Experimental, Universidad de Buenos Aires, Buenos Aires, Argentina.,Instituto de Fisiología, Biología Molecular y Neurociencias, CONICET, Buenos Aires, Argentina
| |
Collapse
|
32
|
Shirazi SN, Friedman AR, Kaufer D, Sakhai SA. Glucocorticoids and the Brain: Neural Mechanisms Regulating the Stress Response. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015. [DOI: 10.1007/978-1-4939-2895-8_10] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
33
|
Schoenfeld TJ, Cameron HA. Adult neurogenesis and mental illness. Neuropsychopharmacology 2015; 40:113-28. [PMID: 25178407 PMCID: PMC4262910 DOI: 10.1038/npp.2014.230] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 08/28/2014] [Accepted: 08/28/2014] [Indexed: 02/07/2023]
Abstract
Several lines of evidence suggest that adult neurogenesis, the production of new neurons in adulthood, may play a role in psychiatric disorders, including depression, anxiety, and schizophrenia. Medications and other treatments for mental disorders often promote the proliferation of new neurons; the time course for maturation and integration of new neurons in circuitry parallels the delayed efficacy of psychiatric therapies; adverse and beneficial experiences similarly affect development of mental illness and neurogenesis; and ablation of new neurons in adulthood alters the behavioral impact of drugs in animal models. At present, the links between adult neurogenesis and depression seem stronger than those suggesting a relationship between new neurons and anxiety or schizophrenia. Yet, even in the case of depression there is currently no direct evidence for a causative role. This article reviews the data relating adult neurogenesis to mental illness and discusses where research needs to head in the future.
Collapse
Affiliation(s)
- Timothy J Schoenfeld
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Heather A Cameron
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA,Section on Neuroplasticity, NIMH, 35 Convent Drive, Building 35/3C915, Bethesda, MD 20892-3718, USA, Tel: +1 301 496 3814, Fax: +1 301 480 4564, E-mail:
| |
Collapse
|
34
|
Joels G, Lamprecht R. Fear memory formation can affect a different memory: fear conditioning affects the extinction, but not retrieval, of conditioned taste aversion (CTA) memory. Front Behav Neurosci 2014; 8:324. [PMID: 25324744 PMCID: PMC4179742 DOI: 10.3389/fnbeh.2014.00324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Accepted: 09/01/2014] [Indexed: 11/22/2022] Open
Abstract
The formation of fear memory to a specific stimulus leads to subsequent fearful response to that stimulus. However, it is not apparent whether the formation of fear memory can affect other memories. We study whether specific fearful experience leading to fear memory affects different memories formation and extinction. We revealed that cued fear conditioning, but not unpaired or naïve training, inhibited the extinction of conditioned taste aversion (CTA) memory that was formed after fear conditioning training in rats. Fear conditioning had no effect on retrieval of CTA memory but specifically impaired its extinction. Extinguished fear memory, after fear extinction training, had no effect on future CTA memory extinction. Fear conditioning had no effect on CTA memory extinction if CTA memory was formed before fear conditioning. Conditioned taste aversion had no effect on fear conditioning memory extinction. We conclude that active cued fear conditioning memory can affect specifically the extinction, but not the formation, of future different memory.
Collapse
Affiliation(s)
- Gil Joels
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa Haifa, Israel
| | - Raphael Lamprecht
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa Haifa, Israel ; Department of Biology, Faculty of Natural Sciences, University of Haifa Haifa, Israel ; Center for Gene Manipulation in the Brain, University of Haifa Haifa, Israel ; Center for Brain and Behavior, University of Haifa Haifa, Israel
| |
Collapse
|
35
|
Chawana R, Alagaili A, Patzke N, Spocter MA, Mohammed OB, Kaswera C, Gilissen E, Bennett NC, Ihunwo AO, Manger PR. Microbats appear to have adult hippocampal neurogenesis, but post-capture stress causes a rapid decline in the number of neurons expressing doublecortin. Neuroscience 2014; 277:724-33. [PMID: 25106130 DOI: 10.1016/j.neuroscience.2014.07.063] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Revised: 07/24/2014] [Accepted: 07/28/2014] [Indexed: 11/24/2022]
Abstract
A previous study investigating potential adult hippocampal neurogenesis in microchiropteran bats failed to reveal a strong presence of this neural trait. As microchiropterans have a high field metabolic rate and a small body mass, it is possible that capture/handling stress may lead to a decrease in the detectable presence of adult hippocampal neurogenesis. Here we looked for evidence of adult hippocampal neurogenesis using immunohistochemical techniques for the endogenous marker doublecortin (DCX) in 10 species of microchiropterans euthanized and perfusion fixed at specific time points following capture. Our results reveal that when euthanized and perfused within 15 min of capture, abundant putative adult hippocampal neurogenesis could be detected using DCX immunohistochemistry. Between 15 and 30 min post-capture, the detectable levels of DCX dropped dramatically and after 30 min post-capture, immunohistochemistry for DCX could not reveal any significant evidence of putative adult hippocampal neurogenesis. Thus, as with all other mammals studied to date apart from cetaceans, bats, including both microchiropterans and megachiropterans, appear to exhibit substantial levels of adult hippocampal neurogenesis. The present study underscores the concept that, as with laboratory experiments, studies conducted on wild-caught animals need to be cognizant of the fact that acute stress (capture/handling) may induce major changes in the appearance of specific neural traits.
Collapse
Affiliation(s)
- R Chawana
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193 Johannesburg, South Africa
| | - A Alagaili
- KSU Mammals Research Chair, Department of Zoology, College of Sciences, King Saud University, Box 2455, Riyadh 11451, Saudi Arabia
| | - N Patzke
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193 Johannesburg, South Africa
| | - M A Spocter
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193 Johannesburg, South Africa; Department of Anatomy, Des Moines University, Des Moines, Iowa, USA
| | - O B Mohammed
- KSU Mammals Research Chair, Department of Zoology, College of Sciences, King Saud University, Box 2455, Riyadh 11451, Saudi Arabia
| | - C Kaswera
- Faculté des Sciences, University of Kisangani, B.P 1232 Kisangani, Congo
| | - E Gilissen
- Department of African Zoology, Royal Museum for Central Africa, Leuvensesteenweg 13, B-3080 Tervuren, Belgium; Laboratory of Histology and Neuropathology, Université Libre de Bruxelles, 1070 Brussels, Belgium; Department of Anthropology, University of Arkansas, Fayetteville, AR 72701, USA
| | - N C Bennett
- Department of Zoology and Entomology, University of Pretoria, Pretoria 0002, South Africa
| | - A O Ihunwo
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193 Johannesburg, South Africa
| | - P R Manger
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, 2193 Johannesburg, South Africa.
| |
Collapse
|
36
|
Forced swim and chronic variable stress reduced hippocampal cell survival in OVX female rats. Behav Brain Res 2014; 270:248-55. [DOI: 10.1016/j.bbr.2014.05.033] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Revised: 05/13/2014] [Accepted: 05/16/2014] [Indexed: 12/13/2022]
|
37
|
Further evidence for the role of interferon-gamma on anxiety- and depressive-like behaviors: involvement of hippocampal neurogenesis and NGF production. Neurosci Lett 2014; 578:100-5. [PMID: 24993299 DOI: 10.1016/j.neulet.2014.06.039] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 06/06/2014] [Accepted: 06/23/2014] [Indexed: 11/21/2022]
Abstract
A series of evidence suggests that interferon-gamma (IFN-γ) plays an important role in central nervous system (CNS) functions. However, previous studies have obtained inconsistent results regarding the role of IFN-γ in modulating emotion-related behaviors. The present study aimed to evaluate the behavioral profile of IFN-γ knockout (K.O.) mice in models of anxiety and depression. Male C57Bl6 wild type (WT) or IFN-γ K.O. mice were submitted to the following tests: contextual fear conditioning (CFC), elevated plus maze (EPM), open field (OF) and forced swimming test (FST). To explore the possible neurobiological mechanisms involved, we also assessed hippocampal neurogenesis by means of hippocampal doublecortin expression, and the levels of brain-derived neurothophic factor (BDNF) and nerve growth factor (NGF) in the hippocampus and prefrontal cortex. Our results suggested that IFN-γ K.O. mice exhibited an anxiogenic profile in CFC, EPM and OF tests. In FST, the K.O. group spent more time immobile than the WT group. The number of doublecortin positive cells was reduced in the dentate gyrus, and the expression of NGF was down regulated in the prefrontal cortex of IFN-γ K.O. mice. Our results suggest that IFN-γ is involved in CNS plasticity, contributing to the modulation of anxiety and depressive states.
Collapse
|
38
|
Gao J, Wang H, Liu Y, Li YY, Chen C, Liu LM, Wu YM, Li S, Yang C. Glutamate and GABA imbalance promotes neuronal apoptosis in hippocampus after stress. Med Sci Monit 2014; 20:499-512. [PMID: 24675061 PMCID: PMC3976216 DOI: 10.12659/msm.890589] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background People who experience traumatic events have an increased risk of post-traumatic stress disorder (PTSD). However, PTSD-related pathological changes in the hippocampus and prefrontal cortex remain poorly understood. Material/Methods We investigated the effect of a PTSD-like animal model induced by severe stress. The experimental rats received 20 inescapable electric foot shocks in an enclosed box for a total of 6 times in 3 days. The physiological state (body weight and plasma corticosterone concentrations), emotion, cognitive behavior, brain morphology, apoptosis, and balance of gamma-aminobutyric acid (GABA) and glutamate in the hippocampus and prefrontal cortex were observed. Cell damages were examined with histological staining (HE, Nissl, and silver impregnation), while apoptosis was analyzed with flow cytometry using an Annexin V and propidium iodide (PI) binding and terminal deoxynucleotidyl transferase mediated-dUTP nick end labeling (TUNEL) method. Results In comparison with the sham litter-mates, the stressed rats showed decreased body weight, inhibition of hypothalamic-pituitary-adrenal (HPA) axis activation, increase in freezing response to trauma reminder, hypoactivity and anxiety-like behaviors in elevated plus maze and open field test, poor learning in Morris water maze, and shortened latency in hot-plate test. There were significant damages in the hippocampus but not in the prefrontal cortex. Imbalance between glutamate and GABA was more evident in the hippocampus than in the prefrontal cortex. Conclusions These results suggest that neuronal apoptosis in the hippocampus after severe traumatic stress is related to the imbalance between glutamate and GABA. Such modifications may resemble the profound changes observed in PTSD patients.
Collapse
Affiliation(s)
- Jie Gao
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China (mainland)
| | - He Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China (mainland)
| | - Yuan Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China (mainland)
| | - Ying-Yu Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China (mainland)
| | - Can Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China (mainland)
| | - Liang-Ming Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China (mainland)
| | - Ya-Min Wu
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China (mainland)
| | - Sen Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China (mainland)
| | - Ce Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China (mainland)
| |
Collapse
|
39
|
Monsey MS, Boyle LM, Zhang ML, Nguyen CP, Kronman HG, Ota KT, Duman RS, Taylor JR, Schafe GE. Chronic corticosterone exposure persistently elevates the expression of memory-related genes in the lateral amygdala and enhances the consolidation of a Pavlovian fear memory. PLoS One 2014; 9:e91530. [PMID: 24618807 PMCID: PMC3950278 DOI: 10.1371/journal.pone.0091530] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2014] [Accepted: 02/11/2014] [Indexed: 01/22/2023] Open
Abstract
Chronic exposure to stress has been widely implicated in the development of anxiety disorders, yet relatively little is known about the long-term effects of chronic stress on amygdala-dependent memory formation. Here, we examined the effects of a history of chronic exposure to the stress-associated adrenal steroid corticosterone (CORT) on the consolidation of a fear memory and the expression of memory-related immediate early genes (IEGs) in the lateral nucleus of the amygdala (LA). Rats received chronic exposure to CORT (50 μg/ml) in their drinking water for 2 weeks and were then titrated off the CORT for an additional 6 days followed by a 2 week ‘wash-out’ period consisting of access to plain water. Rats were then either sacrificed to examine the expression of memory-related IEG expression in the LA or given auditory Pavlovian fear conditioning. We show that chronic exposure to CORT leads to a persistent elevation in the expression of the IEGs Arc/Arg3.1 and Egr-1 in the LA. Further, we show that rats with a history of chronic CORT exposure exhibit enhanced consolidation of a fear memory; short-term memory (STM) is not affected, while long-term memory (LTM) is significantly enhanced. Treatment with the selective serotonin reuptake inhibitor (SSRI) fluoxetine following the chronic CORT exposure period was observed to effectively reverse both the persistent CORT-related increases in memory-related IEG expression in the LA and the CORT-related enhancement in fear memory consolidation. Our findings suggest that chronic exposure to CORT can regulate memory-related IEG expression and fear memory consolidation processes in the LA in a long-lasting manner and that treatment with fluoxetine can reverse these effects.
Collapse
Affiliation(s)
- Melissa S. Monsey
- Department of Psychology, Yale University, New Haven, Connecticut, United States of America
| | - Lara M. Boyle
- Department of Psychology, Yale University, New Haven, Connecticut, United States of America
| | - Melinda L. Zhang
- Department of Psychology, Yale University, New Haven, Connecticut, United States of America
| | - Caroline P. Nguyen
- Department of Psychology, Yale University, New Haven, Connecticut, United States of America
| | - Hope G. Kronman
- Department of Psychology, Yale University, New Haven, Connecticut, United States of America
| | - Kristie T. Ota
- Department of Psychiatry, Yale University, New Haven, Connecticut, United States of America
| | - Ronald S. Duman
- Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut, United States of America
- Department of Psychiatry, Yale University, New Haven, Connecticut, United States of America
| | - Jane R. Taylor
- Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut, United States of America
- Department of Psychiatry, Yale University, New Haven, Connecticut, United States of America
| | - Glenn E. Schafe
- Department of Psychology, The City University of New York, New York, New York, United States of America
- Center for Study of Gene Structure and Function, Hunter College, The City University of New York, New York, New York, United States of America
- The Graduate Center, The City University of New York, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
40
|
Sántha P, Pákáski M, Fodor EK, Fazekas ÖC, Kálmán S, Kálmán J, Janka Z, Szabó G, Kálmán J. Cytoskeletal protein translation and expression in the rat brain are stressor-dependent and region-specific. PLoS One 2013; 8:e73504. [PMID: 24124448 PMCID: PMC3790765 DOI: 10.1371/journal.pone.0073504] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 07/23/2013] [Indexed: 11/19/2022] Open
Abstract
Stress is an integral component of life that can sometimes cause a critical overload, depending on the qualitative and quantitative natures of the stressors. The involvement of actin, the predominant component of dendritic integrity, is a plausible candidate factor in stress-induced neuronal cytoskeletal changes. The major aim of this study was to compare the effects of three different stress conditions on the transcription and translation of actin-related cytoskeletal genes in the rat brain. Male Wistar rats were exposed to one or other of the frequently used models of physical stress, i.e. electric foot shock stress (EFSS), forced swimming stress (FSS), or psychosocial stress (PSS) for periods of 3, 7, 14, or 21 days. The relative mRNA and protein expressions of β-actin, cofilin and mitogen-activated protein kinase 1 (MAPK-1) were determined by qRT- PCR and western blotting from hippocampus and frontal cortex samples. Stressor-specific alterations in both β-actin and cofilin expression levels were seen after stress. These alterations were most pronounced in response to EFSS, and exhibited a U-shaped time course. FSS led to a significant β-actin mRNA expression elevation in the hippocampus and the frontal cortex after 3 and 7 days, respectively, without any subsequent change. PSS did not cause any change in β-actin or cofilin mRNA or protein expression in the examined brain regions. EFSS, FSS and PSS had no effect on the expression of MAPK-1 mRNA at any tested time point. These findings indicate a very delicate, stress type-dependent regulation of neuronal cytoskeletal components in the rat hippocampus and frontal cortex.
Collapse
Affiliation(s)
- Petra Sántha
- Alzheimer's Disease Research Centre, Department of Psychiatry, University of Szeged, Szeged, Hungary
- * E-mail:
| | - Magdolna Pákáski
- Alzheimer's Disease Research Centre, Department of Psychiatry, University of Szeged, Szeged, Hungary
| | - Eszter K. Fodor
- Alzheimer's Disease Research Centre, Department of Psychiatry, University of Szeged, Szeged, Hungary
| | - Örsike Cs Fazekas
- Alzheimer's Disease Research Centre, Department of Psychiatry, University of Szeged, Szeged, Hungary
| | - Sára Kálmán
- Alzheimer's Disease Research Centre, Department of Psychiatry, University of Szeged, Szeged, Hungary
| | - János Kálmán
- Alzheimer's Disease Research Centre, Department of Psychiatry, University of Szeged, Szeged, Hungary
| | - Zoltán Janka
- Department of Psychiatry, University of Szeged, Szeged, Hungary
| | - Gyula Szabó
- Department of Pathophysiology, University of Szeged, Szeged, Hungary
| | - János Kálmán
- Alzheimer's Disease Research Centre, Department of Psychiatry, University of Szeged, Szeged, Hungary
| |
Collapse
|
41
|
Kuipers SD, Trentani A, van der Zee EA, den Boer JA. Chronic stress-induced changes in the rat brain: role of sex differences and effects of long-term tianeptine treatment. Neuropharmacology 2013; 75:426-36. [PMID: 23994757 DOI: 10.1016/j.neuropharm.2013.08.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 08/13/2013] [Accepted: 08/15/2013] [Indexed: 01/28/2023]
Abstract
Growing evidence suggests neuroplasticity changes are pivotal in both the occurrence and treatment of affective disorders. Abnormal expression and/or phosphorylation of numerous plasticity-related proteins have been observed in depression, while prolonged antidepressant treatment has been associated with the attenuation of stress-mediated effects on dendritic remodeling and adult hippocampal neurogenesis in experimental animals. This study explores the neurobiological adaptations induced by chronic stress and/or long-term tianeptine treatment. Male and female rats were studied to determine the potential contributory role of sex differences on stress-induced pathology and antidepressant-mediated actions. Our results confirm chronic stress-induced HPA axis disturbance and neuroplasticity impairment in both sexes (i.e. reduced CREB phosphorylation and hippocampal BrdU labeling). Commonly ensuing neurobiological alterations were accompanied by unique sex-specific adaptations. When the antidepressant tianeptine was administered, HPA axis hyperactivity was attenuated and specific neuronal defects were ameliorated in both sexes. These findings provide novel insight into sex-related influences on the neurobiological substrates mediating chronic stress-induced actions on neuroplasticity and the mechanisms underlying tianeptine-mediated therapeutic effects.
Collapse
Affiliation(s)
- Sjoukje D Kuipers
- University of Groningen, University Medical Centre Groningen, Department of Psychiatry, The Netherlands; Department of Biomedicine, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway
| | - Andrea Trentani
- University of Groningen, University Medical Centre Groningen, Department of Psychiatry, The Netherlands; Department of Molecular Neurobiology, Centre for Behaviour and Neurosciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands; Department of Biomedicine, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway.
| | - Eddy A van der Zee
- Department of Molecular Neurobiology, Centre for Behaviour and Neurosciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Johan A den Boer
- University of Groningen, University Medical Centre Groningen, Department of Psychiatry, The Netherlands
| |
Collapse
|
42
|
Green MR, McCormick CM. Effects of stressors in adolescence on learning and memory in rodent models. Horm Behav 2013; 64:364-79. [PMID: 23998678 DOI: 10.1016/j.yhbeh.2012.09.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 09/04/2012] [Accepted: 09/23/2012] [Indexed: 02/07/2023]
Abstract
This article is part of a Special Issue "Puberty and Adolescence". Learning and memory is affected by a myriad of factors, including exposure to stressors and the corresponding rise in circulating glucocorticoids. Nevertheless, the effects of stressors depend on the sex, species, the type of stressor used, the duration of exposure, as well as the developmental time-point in which stressors are experienced. Effects of stress in adolescence, however, have received less attention than other developmental periods. In adolescence, the hypothalamic-pituitary-adrenal axis and brain regions involved in learning and memory, which also richly express corticosteroid receptors, are continuing to develop, and thus the effects of stress exposures would be expected to differ from those in adulthood. We conclude from a review of the available literature in animal models that hippocampal function is particularly sensitive to adolescent stressors, and the effects tend to be most evident several weeks after the exposure, suggesting stressors alter the developmental trajectory of the hippocampus.
Collapse
Affiliation(s)
- Matthew R Green
- Department of Psychology, Brock University, 500 Glenridge Ave., St. Catharines, Ontario, Canada
| | | |
Collapse
|
43
|
Kirby ED, Muroy SE, Sun WG, Covarrubias D, Leong MJ, Barchas LA, Kaufer D. Acute stress enhances adult rat hippocampal neurogenesis and activation of newborn neurons via secreted astrocytic FGF2. eLife 2013; 2:e00362. [PMID: 23599891 PMCID: PMC3628086 DOI: 10.7554/elife.00362] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 02/27/2013] [Indexed: 12/19/2022] Open
Abstract
Stress is a potent modulator of the mammalian brain. The highly conserved stress hormone response influences many brain regions, particularly the hippocampus, a region important for memory function. The effect of acute stress on the unique population of adult neural stem/progenitor cells (NPCs) that resides in the adult hippocampus is unclear. We found that acute stress increased hippocampal cell proliferation and astrocytic fibroblast growth factor 2 (FGF2) expression. The effect of acute stress occurred independent of basolateral amygdala neural input and was mimicked by treating isolated NPCs with conditioned media from corticosterone-treated primary astrocytes. Neutralization of FGF2 revealed that astrocyte-secreted FGF2 mediated stress-hormone-induced NPC proliferation. 2 weeks, but not 2 days, after acute stress, rats also showed enhanced fear extinction memory coincident with enhanced activation of newborn neurons. Our findings suggest a beneficial role for brief stress on the hippocampus and improve understanding of the adaptive capacity of the brain. DOI:http://dx.doi.org/10.7554/eLife.00362.001.
Collapse
Affiliation(s)
- Elizabeth D Kirby
- Helen Wills Neuroscience Institute , University of California, Berkeley , Berkeley , United States
| | | | | | | | | | | | | |
Collapse
|
44
|
Schoenfeld TJ, Gould E. Differential effects of stress and glucocorticoids on adult neurogenesis. Curr Top Behav Neurosci 2013; 15:139-164. [PMID: 23670817 DOI: 10.1007/7854_2012_233] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Stress is known to inhibit neuronal growth in the hippocampus. In addition to reducing the size and complexity of the dendritic tree, stress and elevated glucocorticoid levels are known to inhibit adult neurogenesis. Despite the negative effects of stress hormones on progenitor cell proliferation in the hippocampus, some experiences which produce robust increases in glucocorticoid levels actually promote neuronal growth. These experiences, including running, mating, enriched environment living, and intracranial self-stimulation, all share in common a strong hedonic component. Taken together, the findings suggest that rewarding experiences buffer progenitor cells in the dentate gyrus from the negative effects of elevated stress hormones. This chapter considers the evidence that stress and glucocorticoids inhibit neuronal growth along with the paradoxical findings of enhanced neuronal growth under rewarding conditions with a view toward understanding the underlying biological mechanisms.
Collapse
Affiliation(s)
- Timothy J Schoenfeld
- Department of Psychology, Neuroscience Institute, Princeton University, Princeton, NJ, 08545, USA
| | | |
Collapse
|
45
|
McCormick CM, Green MR, Cameron NM, Nixon F, Levy MJ, Clark RA. Deficits in male sexual behavior in adulthood after social instability stress in adolescence in rats. Horm Behav 2013; 63:5-12. [PMID: 23174754 DOI: 10.1016/j.yhbeh.2012.11.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Revised: 11/04/2012] [Accepted: 11/09/2012] [Indexed: 01/18/2023]
Abstract
There is increasing evidence that exposure to stressors in adolescence has long-lasting effects on emotional and cognitive behavior, but little is known as to whether reproductive functions are affected. We investigated appetitive and consummatory aspects of sexual behavior in male rats that were exposed to chronic social instability stress (SS, n=24) for 16 days in mid-adolescence compared to control rats (CTL, n=24). Over five sexual behavior test sessions with a receptive female, SS rats made fewer ejaculations (p=0.02) and had longer latencies to ejaculation (p=0.03). When only data from rats that ejaculated in the fifth session were analyzed, SS rats (n=18) had reduced copulatory efficiency (more mounts and intromissions before ejaculation) compared to CTL rats (n=19) (p=0.004), and CTL rats were twice as likely as SS rats to make more than one ejaculation in the fifth session (p=0.05). Further, more CTL (14/24) than SS (5/25) rats ejaculated in four or more sessions (p=0.05). SS rats had lower plasma testosterone concentrations than CTL rats (p=0.05), but did not differ in androgen receptor, estrogen receptor alpha, or Fos immunoreactive cell counts in the medial preoptic area. The groups did not differ in a partner preference test administered between the fourth and fifth sexual behavior session. The results suggest that developmental history contributes to individual differences in reproductive behavior, and that stress exposures in adolescence may be a factor in sexual sluggishness.
Collapse
Affiliation(s)
- Cheryl M McCormick
- Department of Psychology, 500 Glenridge Ave., St. Catharines, Ontario, Canada L2S 3A1.
| | | | | | | | | | | |
Collapse
|
46
|
Despite higher glucocorticoid levels and stress responses in female rats, both sexes exhibit similar stress-induced changes in hippocampal neurogenesis. Behav Brain Res 2012; 234:357-64. [DOI: 10.1016/j.bbr.2012.07.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 06/29/2012] [Accepted: 07/07/2012] [Indexed: 11/17/2022]
|
47
|
Early-life stress mediated modulation of adult neurogenesis and behavior. Behav Brain Res 2012; 227:400-9. [DOI: 10.1016/j.bbr.2011.07.037] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Revised: 07/18/2011] [Accepted: 07/21/2011] [Indexed: 02/06/2023]
|
48
|
Regular exercise cures depression-like behavior via VEGF-Flk-1 signaling in chronically stressed mice. Neuroscience 2012; 207:208-17. [PMID: 22306286 DOI: 10.1016/j.neuroscience.2012.01.023] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Revised: 12/26/2011] [Accepted: 01/06/2012] [Indexed: 12/25/2022]
Abstract
In animals, chronic stress leads to the development of depression-like behavior and decreases neurogenesis and blood vessel density in hippocampus, whereas antidepressants increase adult neurogenesis in hippocampus. Regular exercise training also has antidepressant action and increases hippocampal neurogenesis; however, whether exercise-induced antidepressant action is related to hippocampal microvasculature is unclear. To address this issue, we compared depression-like behavior, blood vessel density, and neurogenesis in hippocampal dentate gyrus between stressed and exercised mice with or without administration of inhibitor of vascular endothelial growth factor (VEGF) receptor. Chronic stress led to the development of depression-like behavior, decreased blood vessel density, and neurogenesis in hippocampus. Regular exercise training improved depression-like behavior, the decrease of hippocampal blood vessel density, and neurogenesis in the stress state, whereas the combination of regular exercise and administration of SU1498, VEGF receptor Flk-1 inhibitor, canceled the exercise-induced antidepressant effect. These findings suggested that the improvement of hippocampal blood vessel and adult neurogenesis via VEGF signaling pathway is necessary for exercise-induced antidepressant effect.
Collapse
|
49
|
Zhao Y, Wang Z, Dai J, Chen L, Huang Y, Zhan Z. Beneficial effects of benzodiazepine diazepam on chronic stress-induced impairment of hippocampal structural plasticity and depression-like behavior in mice. Behav Brain Res 2011; 228:339-50. [PMID: 22198054 DOI: 10.1016/j.bbr.2011.12.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 12/03/2011] [Accepted: 12/06/2011] [Indexed: 01/20/2023]
Abstract
Whether benzodiazepines (BZDs) have beneficial effects on the progress of chronic stress-induced impairment of hippocampal structural plasticity and major depression is uncertain. The present study designed four preclinical experiments to determine the effects of BZDs using chronic unpredictable stress model. In Experiment 1, several time course studies on behavior and hippocampus response to stress were conducted using the forced swim and tail suspension tests (FST and TST) as well as hippocampal structural plasticity markers. Chronic stress induced depression-like behavior in the FST and TST as well as decreased hippocampal structural plasticity that returned to normal within 3 wk. In Experiment 2, mice received p.o. administration of three diazepam dosages prior to each variate stress session for 4 wk. This treatment significantly antagonized the elevation of stress-induced corticosterone levels. Only low- (0.5mg/kg) and medium-dose (1mg/kg) diazepam blocked the detrimental effects of chronic stress. In Experiment 3, after 7 wk of stress sessions, daily p.o. diazepam administration during 1 wk recovery phase dose-dependently accelerated the recovery of stressed mice. In Experiment 4, 1 wk diazepam administration to control mice enhanced significantly hippocampal structural plasticity and induced an antidepressant-like behavioral effect, whereas 4 wk diazepam administration produced opposite effects. Hence, diazepam can slow the progress of chronic stress-induced detrimental consequences by normalizing glucocorticoid hormones. Considering the adverse effect of long-term diazepam administration on hippocampal plasticity, the preventive effects of diazepam may depend on the proper dose. Short-term diazepam treatment enhances hippocampal structural plasticity and is beneficial to recovery following chronic stress.
Collapse
Affiliation(s)
- Yunan Zhao
- Key Laboratory of Brain Research, Basic Medical College, Nanjing University of Traditional Chinese Medicine, Nanjing, China.
| | | | | | | | | | | |
Collapse
|
50
|
Hanson ND, Owens MJ, Nemeroff CB. Depression, antidepressants, and neurogenesis: a critical reappraisal. Neuropsychopharmacology 2011; 36:2589-602. [PMID: 21937982 PMCID: PMC3230505 DOI: 10.1038/npp.2011.220] [Citation(s) in RCA: 214] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The neurogenesis hypothesis of depression posits (1) that neurogenesis in the subgranular zone of the dentate gyrus is regulated negatively by stressful experiences and positively by treatment with antidepressant drugs and (2) that alterations in the rate of neurogenesis play a fundamental role in the pathology and treatment of major depression. This hypothesis is supported by important experimental observations, but is challenged by equally compelling contradictory reports. This review summarizes the phenomenon of adult hippocampal neurogenesis, the initial and continued evidence leading to the development of the neurogenesis hypothesis of depression, and the recent studies that have disputed and/or qualified those findings, to conclude that it can be affected by stress and antidepressants under certain conditions, but that these effects do not appear in all cases of psychological stress, depression, and antidepressant treatment.
Collapse
Affiliation(s)
- Nicola D Hanson
- Laboratory of Neuropsychopharmacology, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Michael J Owens
- Laboratory of Neuropsychopharmacology, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA,Laboratory of Neuropsychopharmacology, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, 100 Woodruff Circle, Suite 4000, Atlanta, GA 30322, USA. Tel: +1 404 727 4059, Fax: +1 404 727 3233, E-mail:
| | - Charles B Nemeroff
- Department of Psychiatry and Behavioral Sciences, University of Miami School of Medicine, Miami, FL, USA
| |
Collapse
|