1
|
Popovičová A, Račeková E, Martončíková M, Fabianová K, Raček A, Žideková M. Effect of microwave radiation on adult neurogenesis and behavior of prenatally exposed rats. IBRO Neurosci Rep 2024; 17:235-244. [PMID: 39286040 PMCID: PMC11404077 DOI: 10.1016/j.ibneur.2024.08.007] [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: 04/11/2024] [Revised: 07/31/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Abstract
Postnatal neurogenesis appears to be highly sensitive to environmental factors, including microwave electromagnetic radiation (MWR). Here, we investigated the impact of MWR during intrauterine development on juvenile and adult neurogenesis in the rostral migratory stream (RMS) and the dentate gyrus of the hippocampus in the rat brain, as well as its effect on animal behavior. Female rats were exposed to MWR at a frequency of 2.45 GHz for 2 hours daily throughout pregnancy. The offspring of irradiated mothers survived to either juvenile age or adulthood. The brains of the rats were subjected to morphological analysis, assessing cell proliferation and death in both neurogenic regions. In the RMS, the differentiation of nitrergic neurons was also investigated. The effect of MWR on behavior was evaluated in rats surviving to adulthood. Prenatal MWR exposure caused significant changes in the number of proliferating and dying cells, depending on the age of the animals and the observed neurogenic region. In addition, MWR attenuated the maturation of nitrergic neurons in the RMS in both juvenile and adult rats. Morphological alterations in neurogenesis were accompanied by changes in animals' behavior. Affected neurogenesis and changes in animal behavior suggest a high sensitivity of the developing brain to MWR.
Collapse
Affiliation(s)
- Alexandra Popovičová
- Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Šoltésovej 4, Košice 040 01, Slovakia
| | - Enikő Račeková
- Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Šoltésovej 4, Košice 040 01, Slovakia
| | - Marcela Martončíková
- Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Šoltésovej 4, Košice 040 01, Slovakia
| | - Kamila Fabianová
- Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Šoltésovej 4, Košice 040 01, Slovakia
| | - Adam Raček
- Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Šoltésovej 4, Košice 040 01, Slovakia
| | - Monika Žideková
- Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Šoltésovej 4, Košice 040 01, Slovakia
| |
Collapse
|
2
|
Frazer NB, Kaas GA, Firmin CG, Gamazon ER, Hatzopoulos AK. BMP Antagonist Gremlin 2 Regulates Hippocampal Neurogenesis and Is Associated with Seizure Susceptibility and Anxiety. eNeuro 2024; 11:ENEURO.0213-23.2024. [PMID: 39349059 PMCID: PMC11493175 DOI: 10.1523/eneuro.0213-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 10/02/2024] Open
Abstract
The Bone Morphogenetic Protein (BMP) signaling pathway is vital in neural progenitor cell proliferation, specification, and differentiation. The BMP signaling antagonist Gremlin 2 (Grem2) is the most potent natural inhibitor of BMP expressed in the adult brain; however its function remains unknown. To address this knowledge gap, we have analyzed mice lacking Grem2 via homologous recombination (Grem2-/- ). Histological analysis of brain sections revealed significant scattering of CA3 pyramidal cells within the dentate hilus in the hippocampus of Grem2-/- mice. Furthermore, the number of proliferating neural stem cells and neuroblasts was significantly decreased in the subgranular zone of Grem2-/- mice compared with that of wild-type (WT) controls. Due to the role of hippocampal neurogenesis in neurological disorders, we tested mice on a battery of neurobehavioral tests. Grem2-/- mice exhibited increased anxiety on the elevated zero maze in response to acute and chronic stress. Specifically, male Grem2-/- mice showed increased anxiogenesis following chronic stress, and this was correlated with higher levels of BMP signaling and decreased proliferation in the dentate gyrus. Additionally, when chemically challenged with kainic acid, Grem2-/- mice displayed a higher susceptibility to and increased severity of seizures compared with WTs. Together, our data indicate that Grem2 regulates BMP signaling and is vital in maintaining homeostasis in adult hippocampal neurogenesis and structure. Furthermore, the lack of Grem2 contributes to the development and progression of neurogenesis-related disorders such as anxiety and epilepsy.
Collapse
Affiliation(s)
- Nicolette B Frazer
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232
| | - Garrett A Kaas
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Caroline G Firmin
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Eric R Gamazon
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Antonis K Hatzopoulos
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, Tennessee 37232
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| |
Collapse
|
3
|
Ferreira AC, Marques F. The Effects of Stress on Hippocampal Neurogenesis and Behavior in the Absence of Lipocalin-2. Int J Mol Sci 2023; 24:15537. [PMID: 37958520 PMCID: PMC10649401 DOI: 10.3390/ijms242115537] [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: 09/07/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023] Open
Abstract
Lipocalin-2 (LCN2) is an acute phase protein able to bind iron when complexed with bacterial siderophores. The recent identification of a mammalian siderophore also suggested a physiological role for LCN2 in the regulation of iron levels and redox state. In the central nervous system, the deletion of LCN2 induces deficits in neural stem cells proliferation and commitment, with an impact on the hippocampal-dependent contextual fear discriminative task. Additionally, stress is a well-known regulator of cell genesis and is known to decrease adult hippocampal cell proliferation and neurogenesis. Although voluntary running, another well-known regulator of neurogenesis, is sufficient to rescue the defective hippocampal neurogenesis and behavior in LCN2-null mice by promoting stem cells' cell cycle progression and maturation, the relevance of LCN2-regulated hippocampal neurogenesis in response to stress has never been explored. Here, we show a lack of response by LCN2-null mice to the effects of chronic stress exposure at the cellular and behavioral levels. Together, these findings implicate LCN2 as a relevant mediator of neuronal plasticity and brain function in the adult mammalian brain.
Collapse
Affiliation(s)
- Ana Catarina Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal;
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Fernanda Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal;
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| |
Collapse
|
4
|
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: 2.3] [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
|
5
|
Endogenous Estrogen Influences Predator Odor-Induced Impairment of Cognitive and Social Behaviors in Aromatase Gene Deficiency Mice. Behav Neurol 2021; 2021:5346507. [PMID: 34594430 PMCID: PMC8478571 DOI: 10.1155/2021/5346507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/28/2021] [Indexed: 11/17/2022] Open
Abstract
Epidemiological studies have suggested that traumatic stress increases vulnerability to various mental disorders, such as dementia and psychiatric disorders. While women are more vulnerable than men to depression and anxiety, it is unclear whether endogenous estrogens are responsible for the underlying sex-specific mechanisms. In this study, the aromatase gene heterozygous (Ar+/-) mice were used as an endogenous estrogen deficiency model and age- and sex-matched wild type mice (WT) as controls to study the predator odor 2,3,5-trimethyl-3-thiazoline- (TMT-) induced short- and long-term cognitive and social behavior impairments. In addition, the changes in brain regional neurotransmitters and their associations with TMT-induced changes in behaviors were further investigated in these animals. Our results showed TMT induced immediate fear response in both Ar+/- and WT mice regardless of sexes. TMT induced an acute impairment of novel object recognition memory and long-term social behavior impairment in WT mice, particularly in females, while Ar+/- mice showed impaired novel object recognition in both sexes and TMT-elevated social behaviors, particularly in males. TMT failed to induce changes in the prepulse inhibition (PPI) test in both groups. TMT resulted in a slight increase of DOPAC/DA ratio in the cortex and a significant elevation of this ratio in the striatum of WT mice. In addition, the ratio of HIAA/5-HT was significantly elevated in the cortex of TMT-treated WT mice, which was not found in TMT-treated Ar+/- mice. Taken together, our results indicate that TMT exposure can cause cognitive and social behavior impairments as well as change catecholamine metabolism in WT mice, and endogenous estrogen deficiency might desensitize the behavioral and neurochemical responses to TMT in Ar+/- mice.
Collapse
|
6
|
Moazzami K, Wittbrodt MT, Lima BB, Kim JH, Hammadah M, Ko YA, Obideen M, Abdelhadi N, Kaseer B, Gafeer MM, Nye JA, Shah AJ, Ward L, Raggi P, Waller EK, Bremner JD, Quyyumi AA, Vaccarino V. Circulating Progenitor Cells and Cognitive Impairment in Men and Women with Coronary Artery Disease. J Alzheimers Dis 2021; 74:659-668. [PMID: 32083582 DOI: 10.3233/jad-191063] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Circulating progenitor cells (CPC) have been associated with memory function and cognitive impairment in healthy adults. However, it is unclear whether such associations also exist in patients with coronary artery disease (CAD). OBJECTIVE To assess the association between CPCs and memory performance among individuals with CAD. METHODS We assessed cognitive function in 509 patients with CAD using the verbal and visual Memory subtests of the Wechsler memory scale-IV and the Trail Making Test parts A and B. CPCs were enumerated with flow cytometry as CD45med/CD34+ blood mononuclear cells, those co-expressing other epitopes representing populations enriched for hematopoietic and endothelial progenitors. RESULTS After adjusting for demographic and cardiovascular risk factors, lower number of endothelial progenitor cell counts were independently associated with lower visual and verbal memory scores (p for all < 0.05). There was a significant interaction in the magnitude of this association with race (p < 0.01), such that the association of verbal memory scores with endothelial progenitor subsets was present in Black but not in non-Black participants. No associations were present with the hematopoietic progenitor-enriched cells or with the Trail Making Tests. CONCLUSION Lower numbers of circulating endothelial progenitor cells are associated with cognitive impairment in patients with CAD, suggesting a protective effect of repair/regeneration processes in the maintenance of cognitive status. Impairment of verbal memory function was more strongly associated with lower CPC counts in Black compared to non-Black participants with CAD. Whether strategies designed to improve regenerative capacity will improve cognition needs further study.
Collapse
Affiliation(s)
- Kasra Moazzami
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.,Department of Medicine, Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Matthew T Wittbrodt
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Bruno B Lima
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.,Department of Medicine, Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jeong Hwan Kim
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.,Department of Medicine, Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Muhammad Hammadah
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.,Department of Medicine, Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Yi-An Ko
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Malik Obideen
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.,Department of Medicine, Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Naser Abdelhadi
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.,Department of Medicine, Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Belal Kaseer
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.,Department of Medicine, Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - M Mazen Gafeer
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.,Department of Medicine, Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jonathon A Nye
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA
| | - Amit J Shah
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.,Department of Medicine, Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA.,Atlanta VA Medical Center, Decatur, GA, USA
| | - Laura Ward
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Paolo Raggi
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.,Mazankowski Alberta Heart Institute, University of Alberta, Alberta, Canada
| | - Edmund K Waller
- Department of Hematology and Oncology, Winship Cancer Institute, Atlanta, GA, USA
| | - J Douglas Bremner
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA.,Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, USA.,Atlanta VA Medical Center, Decatur, GA, USA
| | - Arshed A Quyyumi
- Department of Medicine, Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Viola Vaccarino
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA.,Department of Medicine, Emory Clinical Cardiovascular Research Institute, Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
7
|
Conley MI, Skalaban LJ, Rapuano KM, Gonzalez R, Laird AR, Dick AS, Sutherland MT, Watts R, Casey B. Altered hippocampal microstructure and function in children who experienced Hurricane Irma. Dev Psychobiol 2021; 63:864-877. [PMID: 33325561 PMCID: PMC8206237 DOI: 10.1002/dev.22071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/31/2020] [Accepted: 11/25/2020] [Indexed: 01/12/2023]
Abstract
Hurricane Irma was the most powerful Atlantic hurricane in recorded history, displacing 6 million and killing over 120 people in the state of Florida alone. Unpredictable disasters like Irma are associated with poor cognitive and health outcomes that can disproportionately impact children. This study examined the effects of Hurricane Irma on the hippocampus and memory processes previously related to unpredictable stress. We used an innovative application of an advanced diffusion-weighted imaging technique, restriction spectrum imaging (RSI), to characterize hippocampal microstructure (i.e., cell density) in 9- to 10-year-old children who were exposed to Hurricane Irma relative to a non-exposed control group (i.e., assessed the year before Hurricane Irma). We tested the hypotheses that the experience of Hurricane Irma would be associated with decreases in: (a) hippocampal cellularity (e.g., neurogenesis), based on known associations between unpredictable stress and hippocampal alterations; and (b) hippocampal-related memory function as indexed by delayed recall. We show an association between decreased hippocampal cellularity and delayed recall memory in children who experienced Hurricane Irma relative to those who did not. These findings suggest an important role of RSI for assessing subtle microstructural changes related to functionally significant changes in the developing brain in response to environmental events.
Collapse
Affiliation(s)
- May I. Conley
- Department of PsychologyYale UniversityNew HavenCTUSA
| | | | | | - Raul Gonzalez
- Department of PsychologyFlorida International UniversityMiamiFLUSA
| | - Angela R. Laird
- Department of PhysicsFlorida International UniversityMiamiFLUSA
| | | | | | - Richard Watts
- Department of PsychologyYale UniversityNew HavenCTUSA
| | - B.J. Casey
- Department of PsychologyYale UniversityNew HavenCTUSA
| |
Collapse
|
8
|
Zanette LY, Clinchy M. Ecology and Neurobiology of Fear in Free-Living Wildlife. ANNUAL REVIEW OF ECOLOGY, EVOLUTION, AND SYSTEMATICS 2020. [DOI: 10.1146/annurev-ecolsys-011720-124613] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The ecology of fear concerns the population-, community-, and ecosystem-level consequences of the behavioral interactions between predators and prey, i.e., the aggregate impacts of individual responses to life-threatening events. We review new experiments demonstrating that fear itself is powerful enough to affect the population growth rate in free-living wild birds and mammals, and fear of large carnivores—or the human super predator—can cause trophic cascades affecting plant and invertebrate abundance. Life-threatening events like escaping a predator can have enduring, even lifelong, effects on the brain, and new interdisciplinary research on the neurobiology of fear in wild animals is both providing insights into post-traumatic stress (PTSD) and reinforcing the likely commonality of population- and community-level effects of fear in nature. Failing to consider fear thus risks dramatically underestimating the total impact predators can have on prey populations and the critical role predator-prey interactions can play in shaping ecosystems.
Collapse
Affiliation(s)
- Liana Y. Zanette
- Department of Biology, Western University, London, Ontario N6A 5B7, Canada;,
| | - Michael Clinchy
- Department of Biology, Western University, London, Ontario N6A 5B7, Canada;,
| |
Collapse
|
9
|
Kim JL, Bulthuis NE, Cameron HA. The Effects of Anesthesia on Adult Hippocampal Neurogenesis. Front Neurosci 2020; 14:588356. [PMID: 33192273 PMCID: PMC7643675 DOI: 10.3389/fnins.2020.588356] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/22/2020] [Indexed: 01/17/2023] Open
Abstract
In animal studies, prolonged sedation with general anesthetics has resulted in cognitive impairments that can last for days to weeks after exposure. One mechanism by which anesthesia may impair cognition is by decreasing adult hippocampal neurogenesis. Several studies have seen a reduction in cell survival after anesthesia in rodents with most studies focusing on two particularly vulnerable age windows: the neonatal period and old age. However, the extent to which sedation affects neurogenesis in young adults remains unclear. Adult neurogenesis in the dentate gyrus (DG) was analyzed in male and female rats 24 h after a 4-h period of sedation with isoflurane, propofol, midazolam, or dexmedetomidine. Three different cell populations were quantified: cells that were 1 week or 1 month old, labeled with the permanent birthdate markers EdU or BrdU, respectively, and precursor cells, identified by their expression of the endogenous dividing cell marker proliferating cell nuclear antigen (PCNA) at the time of sacrifice. Midazolam and dexmedetomidine reduced cell proliferation in the adult DG in both sexes but had no effect on postmitotic cells. Propofol reduced the number of relatively mature, 28-day old, neurons specifically in female rats and had no effects on younger cells. Isoflurane had no detectable effects on any of the cell populations examined. These findings show no general effect of sedation on adult-born neurons but demonstrate that certain sedatives do have drug-specific and sex-specific effects. The impacts observed on different cell populations predict that any cognitive effects of these sedatives would likely occur at different times, with propofol producing a rapid but short-lived impairment and midazolam and dexmedetomidine altering cognition after a several week delay. Taken together, these studies lend support to the hypothesis that decreased neurogenesis in the young adult DG may mediate the effects of sedation on cognitive function.
Collapse
Affiliation(s)
| | | | - Heather A. Cameron
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
10
|
Schoenfeld TJ, Smith JA, Sonti AN, Cameron HA. Adult neurogenesis alters response to an aversive distractor in a labyrinth maze without affecting spatial learning or memory. Hippocampus 2020; 31:102-114. [PMID: 33038042 DOI: 10.1002/hipo.23267] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/02/2020] [Accepted: 09/06/2020] [Indexed: 11/11/2022]
Abstract
Adult neurogenesis has been implicated in learning and memory of complex spatial environments. However, new neurons also play a role in nonmnemonic behavior, including the stress response and attention shifting. Many commonly used spatial tasks are very simple, and unsuitable for detecting neurogenesis effects, or are aversively motivated, making it difficult to dissociate effects on spatial learning and memory from effects on stress. We have therefore created a novel complex spatial environment, the flex maze, to enable reward-mediated testing of spatial learning in a flexibly configurable labyrinth. Using a pharmacogenetic method to completely inhibit neurogenesis in adulthood, we found that rats lacking new neurons (TK rats) and wild type controls completed and remembered most mazes equally well. However, control rats were slower to complete peppermint-scented mazes than other mazes, while neurogenesis-deficient rats showed no effect of mint on maze behavior, completing these mazes significantly faster than control rats. Additional testing found that wild type and TK rats showed similar detection of, avoidance of, and glucocorticoid response to the mint odor. These results suggest that spatial learning and memory in a labyrinth task is unaffected by the loss of new neurons, but that these cells affect the ability of an aversive stimulus to distract rats from completing the maze.
Collapse
Affiliation(s)
- Timothy J Schoenfeld
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Jesse A Smith
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Anup N Sonti
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| | - Heather A Cameron
- Section on Neuroplasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
11
|
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: 1.8] [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
|
12
|
Li A, Yau SY, Machado S, Wang P, Yuan TF, So KF. Enhancement of Hippocampal Plasticity by Physical Exercise as a Polypill for Stress and Depression: A Review. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 18:294-306. [PMID: 30848219 DOI: 10.2174/1871527318666190308102804] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 12/21/2018] [Accepted: 02/10/2019] [Indexed: 12/12/2022]
Abstract
Generation of newborn neurons that form functional synaptic connections in the dentate gyrus of adult mammals, known as adult hippocampal neurogenesis, has been suggested to play critical roles in regulating mood, as well as certain forms of hippocampus-dependent learning and memory. Environmental stress suppresses structural plasticity including adult neurogenesis and dendritic remodeling in the hippocampus, whereas physical exercise exerts opposite effects. Here, we review recent discoveries on the potential mechanisms concerning how physical exercise mitigates the stressrelated depressive disorders, with a focus on the perspective of modulation on hippocampal neurogenesis, dendritic remodeling and synaptic plasticity. Unmasking such mechanisms may help devise new drugs in the future for treating neuropsychiatric disorders involving impaired neural plasticity.
Collapse
Affiliation(s)
- Ang Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Joint International Research Laboratory of CNS Regeneration Ministry of Education, Jinan University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Sergio Machado
- Laboratory of Physical Activity Neuroscience, Physical Activity Sciences Postgraduate Program - Salgado de Oliveira University, Niteroi, Brazil
| | - Pingjie Wang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Joint International Research Laboratory of CNS Regeneration Ministry of Education, Jinan University, Guangzhou, China
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kwok-Fai So
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Joint International Research Laboratory of CNS Regeneration Ministry of Education, Jinan University, Guangzhou, China.,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China.,State Key Laboratory of Brain and Cognitive Sciences, the University of Hong Kong, Hong Kong SAR, China.,Department of Ophthalmology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
13
|
Rose M, Filiatreault A, Guénette J, Williams A, Thomson EM. Ozone increases plasma kynurenine-tryptophan ratio and impacts hippocampal serotonin receptor and neurotrophic factor expression: Role of stress hormones. ENVIRONMENTAL RESEARCH 2020; 185:109483. [PMID: 32278163 DOI: 10.1016/j.envres.2020.109483] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 06/11/2023]
Abstract
Air pollution is associated with adverse impacts on the brain, including cognitive decline and increased incidence of dementia, depression and anxiety; however, underlying mechanisms remain unclear. We have shown that both ozone and particulate matter activate the hypothalamic-pituitary-adrenal (HPA) axis, increasing plasma glucocorticoids and altering mRNA profiles in multiple tissues including the brain. HPA axis dysregulation has been associated with central nervous system impacts, including key effects in the hippocampus; accordingly, we hypothesized that pollutant-dependent increases in glucocorticoid levels impact biological pathways relevant to brain health. Fischer-344 rats were treated with metyrapone (0 or 50 mg/kg), a glucocorticoid synthesis inhibitor, and exposed to ozone (0 or 0.8 ppm) for 4 h (n = 5/group) to investigate the role of glucocorticoids in ozone-dependent effects on tryptophan metabolism and expression of serotonin receptors and neurotrophic factors. Ozone increased plasma levels of the tryptophan metabolite kynurenine (~2-fold) and decreased tryptophan levels (~1.2 fold). Hippocampal expression of serotonin receptors exhibited differential regulation following exposure, and expression of key neurotrophic factors (brain-derived neurotrophic factor, vascular endothelial growth factor A, insulin-like growth factor-1, tyrosine kinase receptor B, b-cell lymphoma 2) was decreased. Some, but not all effects were abrogated by metyrapone treatment, suggesting both glucocorticoid-dependent and -independent regulation. Exposure to exogenous corticosterone (10 mg/kg) followed by clean air reproduced the ozone effects that were blocked with metyrapone, confirming the specificity of effects to glucocorticoids. These results indicate that ozone can modify pathways relevant to brain health and establish a role for the HPA axis in mediating these effects.
Collapse
Affiliation(s)
- Mercedes Rose
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, K1A 0K9, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, K1H 8M5, Canada
| | - Alain Filiatreault
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, K1A 0K9, Canada
| | - Josée Guénette
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, K1A 0K9, Canada
| | - Andrew Williams
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, K1A 0K9, Canada
| | - Errol M Thomson
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, K1A 0K9, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, K1H 8M5, Canada.
| |
Collapse
|
14
|
Dunlap KD, Vergara MM, Corbo JH. Reduced brain cell proliferation following somatic injury is buffered by social interaction in electric fish, Apteronotus leptorhynchus. Dev Neurobiol 2020; 80:168-177. [PMID: 32452106 DOI: 10.1002/dneu.22760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/13/2020] [Accepted: 05/18/2020] [Indexed: 11/12/2022]
Abstract
In many species, the negative effects of aversive stimuli are mitigated by social interactions, a phenomenon termed social buffering. In one form of social buffering, social interactions reduce the inhibition of brain cell proliferation during stress. Indirect predator stimuli (e.g., olfactory or visual cues) are known to decrease brain cell proliferation, but little is known about how somatic injury, as might occur from direct predator encounter, affects brain cell proliferation and whether this response is influenced by conspecific interactions. Here, we assessed the social buffering of brain cell proliferation in an electric fish, Apteronotus leptorhynchus, by examining the separate and combined effects of tail injury and social interactions. We mimicked a predator-induced injury by amputating the caudal tail tip, exposed fish to paired interactions that varied in timing, duration and recovery period, and measured brain cell proliferation and the degree of social affiliation. Paired social interaction mitigated the negative effects of tail amputation on cell proliferation in the forebrain but not the midbrain. Social interaction either before or after tail amputation reduced the effect of tail injury and continuous interaction both before and after caused an even greater buffering effect. Social interaction buffered the proliferation response after short-term (1 d) or long-term recovery (7 d) from tail amputation. This is the first report of social buffering of brain cell proliferation in a non-mammalian model. Despite the positive association between social stimuli and brain cell proliferation, we found no evidence that fish affiliate more closely following tail injury.
Collapse
Affiliation(s)
- Kent D Dunlap
- Department of Biology, Trinity College, Hartford, CT, USA
| | | | - Joshua H Corbo
- Department of Biology, Trinity College, Hartford, CT, USA
| |
Collapse
|
15
|
Wang T, Bai S, Wang W, Chen Z, Chen J, Liang Z, Qi X, Shen H, Xie P. Diterpene Ginkgolides Exert an Antidepressant Effect Through the NT3-TrkA and Ras-MAPK Pathways. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1279-1294. [PMID: 32308365 PMCID: PMC7132272 DOI: 10.2147/dddt.s229145] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 03/16/2020] [Indexed: 12/13/2022]
Abstract
Background Depression is a highly prevalent mental illness that severely impacts the quality of life of affected individuals. Our recent studies demonstrated that diterpene ginkgolides (DG) have antidepressant effects in mice. However, the underlying molecular mechanisms remained much unclear. Methods In this study, we assessed the antidepressant effects of chronic DG therapy in rats by evaluating depression-related behaviors, we also examined potential side effects using biochemical indicators. Furthermore, we performed an in-depth molecular network analysis of gene–protein–metabolite interactions on the basis of metabolomics. Results Chronic DG treatment significantly ameliorated the depressive-like behavioral phenotype. Furthermore, the neurotrophin signaling-related NT3-TrkA and Ras-MAPK pathways may play an important role in the antidepressant effect of DG in the hippocampus. Conclusion These findings provide novel insight into the mechanisms underlying the antidepressant action of DG, and should help advance the development of new therapeutic strategies for depression.
Collapse
Affiliation(s)
- Ting Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Neurobiology, Chongqing, People's Republic of China
| | - Shunjie Bai
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Neurobiology, Chongqing, People's Republic of China
| | - Wei Wang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Neurobiology, Chongqing, People's Republic of China.,Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Zhi Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Neurobiology, Chongqing, People's Republic of China
| | - Jianjun Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Neurobiology, Chongqing, People's Republic of China
| | - Zihong Liang
- Department of Neurology, The Inner Mongolia Autonomous Region People's Hospital, Hohhot, Inner Mongolia, People's Republic of China
| | - Xunzhong Qi
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Neurobiology, Chongqing, People's Republic of China.,Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Hailan Shen
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Peng Xie
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Chongqing Key Laboratory of Neurobiology, Chongqing, People's Republic of China.,Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.,Department of Neurology, The Inner Mongolia Autonomous Region People's Hospital, Hohhot, Inner Mongolia, People's Republic of China.,Chongqing Key Laboratory of Cerebrovascular Disease Research, Chongqing, People's Republic of China
| |
Collapse
|
16
|
Dunlap KD, Corbo JH, Vergara MM, Beston SM, Walsh MR. Predation drives the evolution of brain cell proliferation and brain allometry in male Trinidadian killifish, Rivulus hartii. Proc Biol Sci 2019; 286:20191485. [PMID: 31822257 PMCID: PMC6939915 DOI: 10.1098/rspb.2019.1485] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 11/14/2019] [Indexed: 02/03/2023] Open
Abstract
The external environment influences brain cell proliferation, and this might contribute to brain plasticity underlying adaptive behavioural changes. Additionally, internal genetic factors influence the brain cell proliferation rate. However, to date, researchers have not examined the importance of environmental versus genetic factors in causing natural variation in brain cell proliferation. Here, we examine brain cell proliferation and brain growth trajectories in free-living populations of Trinidadian killifish, Rivulus hartii, exposed to contrasting predation environments. Compared to populations without predators, populations in high predation (HP) environments exhibited higher rates of brain cell proliferation and a steeper brain growth trajectory (relative to body size). To test whether these differences in the wild persist in a common garden environment, we reared first-generation fish originating from both predation environments in uniform laboratory conditions. Just as in the wild, brain cell proliferation and brain growth in the common garden were greater in HP populations than in no predation populations. The differences in cell proliferation observed across the brain in both the field and common garden studies indicate that the differences are probably genetically based and are mediated by evolutionary shifts in overall brain growth and life-history traits.
Collapse
Affiliation(s)
- Kent D. Dunlap
- Department of Biology, Trinity College, Hartford, CT 06106, USA
| | - Joshua H. Corbo
- Department of Biology, Trinity College, Hartford, CT 06106, USA
| | | | - Shannon M. Beston
- Department of Biology, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Matthew R. Walsh
- Department of Biology, University of Texas at Arlington, Arlington, TX 76019, USA
| |
Collapse
|
17
|
McAllister BB, Pochakom A, Fu S, Dyck RH. Effects of social defeat stress and fluoxetine treatment on neurogenesis and behavior in mice that lack zinc transporter 3 (ZnT3) and vesicular zinc. Hippocampus 2019; 30:623-637. [DOI: 10.1002/hipo.23185] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/13/2019] [Accepted: 11/29/2019] [Indexed: 11/12/2022]
Affiliation(s)
- Brendan B. McAllister
- Department of PsychologyUniversity of Calgary Calgary Alberta Canada
- Hotchkiss Brain InstituteUniversity of Calgary Calgary Alberta Canada
| | - Angela Pochakom
- Department of PsychologyUniversity of Calgary Calgary Alberta Canada
- Hotchkiss Brain InstituteUniversity of Calgary Calgary Alberta Canada
| | - Selena Fu
- Department of PsychologyUniversity of Calgary Calgary Alberta Canada
- Hotchkiss Brain InstituteUniversity of Calgary Calgary Alberta Canada
| | - Richard H. Dyck
- Department of PsychologyUniversity of Calgary Calgary Alberta Canada
- Hotchkiss Brain InstituteUniversity of Calgary Calgary Alberta Canada
| |
Collapse
|
18
|
Sun LH, Tzeng WY, Liao YH, Deng WT, Cherng CG, Yu L. Relevance of number and physiological status of conspecifics in preventing stress-induced decreases in newly proliferated cells and neuroblasts. Psychopharmacology (Berl) 2019; 236:3329-3339. [PMID: 31201477 DOI: 10.1007/s00213-019-05290-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 05/30/2019] [Indexed: 12/21/2022]
Abstract
RATIONALE AND OBJECTIVE The presence of three conspecifics prevents stress-induced decreases in newly proliferated cells and neuroblasts in mouse dentate gyrus (DG). In this study, we sought to determine how many conspecifics are required to exert these protective effects against stress. In addition, we manipulated the physiological status of those conspecifics in the context of their stress-buffering effects and used airborne oxytocin exposure as a substitute for the presence of conspecifics. MATERIALS AND METHODS Bromodeoxyuridine staining was used to indicate the newly proliferated cells and co-staining with doublecortin to reveal the proliferative neuroblasts. RESULTS Presentation of three intact and lipopolysaccharide-treated conspecifics prevented the stress-induced decreases in the number of newly proliferated cells and neuroblasts in DG. Presentation of one saline- or oxytocin (OT)-treated conspecific did not exert observable stress-buffering effects. In contrast, airborne oxytocin prevented the stress-induced decreases in DG cell proliferation and early neurogenesis, while pretreatment with L-371,257, a selective OT receptor antagonist, abolished the buffering effects of OT. CONCLUSIONS Physical interaction with the conspecifics and conspecifics' sickness, at best, play a minor role in mediating the buffering effects against stress-induced decreases in DG cell proliferation or early neurogenesis. Moreover, stress-buffering effects are negligible with the presence of only one conspecific. Finally, airborne OT produced stress-buffering effects possibly via its stimulation of OT receptors. Oxytocin merits further study as a substitute for the stress-buffering effects of companions.
Collapse
Affiliation(s)
- Li-Han Sun
- Department of Physiology, National Cheng Kung University College of Medicine, Tainan, 701, Taiwan, Republic of China
| | - Wen-Yu Tzeng
- Department of Physiology, National Cheng Kung University College of Medicine, Tainan, 701, Taiwan, Republic of China
| | - Yi-Han Liao
- Department of Physiology, National Cheng Kung University College of Medicine, Tainan, 701, Taiwan, Republic of China
| | - Wen-Ting Deng
- Department of Physiology, National Cheng Kung University College of Medicine, Tainan, 701, Taiwan, Republic of China
| | - Chianfang G Cherng
- Education Center of Humanities and Social Sciences, National Yang-Ming University, Taipei, 102, Taiwan, Republic of China.
| | - Lung Yu
- Department of Physiology, National Cheng Kung University College of Medicine, Tainan, 701, Taiwan, Republic of China. .,Institute of Behavioral Medicine, National Cheng Kung University College of Medicine, Tainan, 701, Taiwan, Republic of China.
| |
Collapse
|
19
|
Hillerer KM, Slattery DA, Pletzer B. Neurobiological mechanisms underlying sex-related differences in stress-related disorders: Effects of neuroactive steroids on the hippocampus. Front Neuroendocrinol 2019; 55:100796. [PMID: 31580837 PMCID: PMC7115954 DOI: 10.1016/j.yfrne.2019.100796] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/19/2022]
Abstract
Men and women differ in their vulnerability to a variety of stress-related illnesses, but the underlying neurobiological mechanisms are not well understood. This is likely due to a comparative dearth of neurobiological studies that assess male and female rodents at the same time, while human neuroimaging studies often don't model sex as a variable of interest. These sex differences are often attributed to the actions of sex hormones, i.e. estrogens, progestogens and androgens. In this review, we summarize the results on sex hormone actions in the hippocampus and seek to bridge the gap between animal models and findings in humans. However, while effects of sex hormones on the hippocampus are largely consistent in animals and humans, methodological differences challenge the comparability of animal and human studies on stress effects. We summarise our current understanding of the neurobiological mechanisms that underlie sex-related differences in behavior and discuss implications for stress-related illnesses.
Collapse
Affiliation(s)
- Katharina M Hillerer
- Department of Obstetrics and Gynaecology, Salzburger Landeskrankenhaus (SALK), Paracelsus Medical University (PMU), Clinical Research Center Salzburg (CRCS), Salzburg, Austria.
| | - David A Slattery
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital, Goethe University, Frankfurt, Germany
| | - Belinda Pletzer
- Department of Psychology, University of Salzburg, Salzburg, Austria; Centre for Cognitive Neuroscience, University of Salzburg, Salzburg, Austria
| |
Collapse
|
20
|
Matsushita H, Latt HM, Koga Y, Nishiki T, Matsui H. Oxytocin and Stress: Neural Mechanisms, Stress-Related Disorders, and Therapeutic Approaches. Neuroscience 2019; 417:1-10. [PMID: 31400490 DOI: 10.1016/j.neuroscience.2019.07.046] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 12/24/2022]
Abstract
Clinical reports show that oxytocin (OT) is related to stress-related disorders such as depression, anxiety disorder, and post-traumatic stress disorder. Two key structures in the brain should be paid special attention with regard to stress regulation, namely, the hypothalamus and the hippocampus. The former is the region for central command for most, if not all, of the major endocrine systems, and the latter takes a key position in the regulation of mood and anxiety. There are extensive neural projections between the two structures, and both are functionally intertwined. The hypothalamus projects OTergic neurons to the hippocampus, and the latter possesses high levels of OT receptors. The hippocampus also regulates the secretion of glucocorticoids, a major group of stress hormones. Excessive levels of glucocorticoids in chronic stress cause atrophy of the hippocampus, whereas OT has been shown to protect hippocampal neurons from the toxic effects of glucocorticoids. In this article, we discuss how neural and endocrine mechanisms interplay in stress regulation, with an emphasis on the role of OT, as well as its therapeutic potential in the treatment of stress-related disorders.
Collapse
Affiliation(s)
- Hiroaki Matsushita
- Department of Physiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan.
| | - Hein Min Latt
- Department of Physiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan.
| | - Yuuri Koga
- Department of Physiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Teiichi Nishiki
- Department of Physiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| | - Hideki Matsui
- Department of Physiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
| |
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: 1.0] [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
|
Shallcross J, Hámor P, Bechard AR, Romano M, Knackstedt L, Schwendt M. The Divergent Effects of CDPPB and Cannabidiol on Fear Extinction and Anxiety in a Predator Scent Stress Model of PTSD in Rats. Front Behav Neurosci 2019; 13:91. [PMID: 31133832 PMCID: PMC6523014 DOI: 10.3389/fnbeh.2019.00091] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 04/16/2019] [Indexed: 01/12/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) currently has no FDA-approved treatments that reduce symptoms in the majority of patients. The ability to extinguish fear memory associations is impaired in PTSD individuals. As such, the development of extinction-enhancing pharmacological agents to be used in combination with exposure therapies may benefit the treatment of PTSD. Both mGlu5 and CB1 receptors have been implicated in contextual fear extinction. Thus, here we tested the ability of the mGlu5 positive allosteric modulator 3-Cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB) and cannabidiol (CBD) to reduce both conditioned and unconditioned fear. We used a predator-threat animal model of PTSD which we and others have previously shown to capture the heterogeneity of anxiety responses observed in humans exposed to trauma. Here, 1 week following a 10-min exposure to predator scent stress, rats were classified into stress-Susceptible and stress-Resilient phenotypes using behavioral criteria for elevated plus maze and acoustic startle response performance. Two weeks after classification, rats underwent 3 days of contextual fear extinction and were treated with vehicle, CDPPB or CBD prior to each session. Finally, the light-dark box test was employed to assess phenotypic differences and the effects of CDPPB and CBD on unconditioned anxiety. CDPBB but not CBD, reduced freezing in Susceptible rats relative to vehicle. In the light-dark box test for unconditioned anxiety, CBD, but not CDPPB, reduced anxiety in Susceptible rats. Resilient rats displayed reduced anxiety in the light-dark box relative to Susceptible rats. Taken together, the present data indicate that enhancement of mGlu5 receptor signaling in populations vulnerable to stress may serve to offset a resistance to fear memory extinction without producing anxiogenic effects. Furthermore, in a susceptible population, CBD attenuates unconditioned but not conditioned fear. Taken together, these findings support the use of predator-threat stress exposure in combination with stress-susceptibility phenotype classification as a model for examining the unique drug response profiles and altered neuronal function that emerge as a consequence of the heterogeneity of psychophysiological response to stress.
Collapse
Affiliation(s)
- John Shallcross
- Department of Psychology, University of Florida, Gainesville, FL, United States.,Center for Addiction Research & Education, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Peter Hámor
- Department of Psychology, University of Florida, Gainesville, FL, United States.,Center for Addiction Research & Education, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Allison R Bechard
- Department of Psychology, University of Florida, Gainesville, FL, United States
| | - Madison Romano
- Department of Psychology, University of Florida, Gainesville, FL, United States
| | - Lori Knackstedt
- Department of Psychology, University of Florida, Gainesville, FL, United States.,Center for Addiction Research & Education, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Marek Schwendt
- Department of Psychology, University of Florida, Gainesville, FL, United States.,Center for Addiction Research & Education, College of Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
23
|
Validation of hippocampal biomarkers of cumulative affective experience. Neurosci Biobehav Rev 2019; 101:113-121. [PMID: 30951763 PMCID: PMC6525303 DOI: 10.1016/j.neubiorev.2019.03.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 03/29/2019] [Accepted: 03/31/2019] [Indexed: 12/29/2022]
Abstract
Recent knowledge on hippocampal structural plasticity is reviewed. This knowledge is harnessed to develop biomarkers of cumulative experience. Hippocampal plasticity is shown to have construct, content and criterion validity in mammals. The biomarkers require further validation to be used in birds and fish. We discuss some practical considerations to implement the biomarkers.
Progress in improving the welfare of captive animals has been hindered by a lack of objective indicators to assess the quality of lifetime experience, often called cumulative affective experience. Recent developments in stress biology and psychiatry have shed new light on the role of the mammalian hippocampus in affective processes. Here we review these findings and argue that structural hippocampal biomarkers demonstrate criterion, construct and content validity as indicators of cumulative affective experience in mammals. We also briefly review emerging findings in birds and fish, which have promising implications for applying the hippocampal approach to these taxa, but require further validation. We hope that this review will motivate welfare researchers and neuroscientists to explore the potential of hippocampal biomarkers of cumulative affective experience.
Collapse
|
24
|
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: 4.8] [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
|
25
|
Marques BL, Carvalho GA, Freitas EMM, Chiareli RA, Barbosa TG, Di Araújo AGP, Nogueira YL, Ribeiro RI, Parreira RC, Vieira MS, Resende RR, Gomez RS, Oliveira-Lima OC, Pinto MCX. The role of neurogenesis in neurorepair after ischemic stroke. Semin Cell Dev Biol 2019; 95:98-110. [PMID: 30550812 DOI: 10.1016/j.semcdb.2018.12.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 12/19/2022]
Abstract
Stroke consists of an abrupt reduction of cerebral blood flow resulting in hypoxia that triggers an excitotoxicity, oxidative stress, and neuroinflammation. After the ischemic process, neural precursor cells present in the subventricular zone of the lateral ventricle and subgranular zone of the dentate gyrus proliferate and migrate towards the lesion, contributing to the brain repair. The neurogenesis is induced by signal transduction pathways, growth factors, attractive factors for neuroblasts, transcription factors, pro and anti-inflammatory mediators and specific neurotransmissions. However, this endogenous neurogenesis occurs slowly and does not allow a complete restoration of brain function. Despite that, understanding the mechanisms of neurogenesis could improve the therapeutic strategies for brain repair. This review presents the current knowledge about brain repair process after stroke and the perspectives regarding the development of promising therapies that aim to improve neurogenesis and its potential to form new neural networks.
Collapse
Affiliation(s)
- Bruno L Marques
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Gustavo A Carvalho
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Elis M M Freitas
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Raphaela A Chiareli
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Thiago G Barbosa
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Armani G P Di Araújo
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Yanley L Nogueira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Raul I Ribeiro
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Ricardo C Parreira
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Mariana S Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rodrigo R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Renato S Gomez
- Departamento de Cirurgia, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Onésia C Oliveira-Lima
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil
| | - Mauro C X Pinto
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Goiás, Goiânia, Brazil.
| |
Collapse
|
26
|
BK channel deacetylation by SIRT1 in dentate gyrus regulates anxiety and response to stress. Commun Biol 2018; 1:82. [PMID: 30271963 PMCID: PMC6123630 DOI: 10.1038/s42003-018-0088-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 05/30/2018] [Indexed: 11/09/2022] Open
Abstract
Previous genomic studies in humans indicate that SIRT1, a nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylase, is involved in anxiety and depression, but the mechanisms are unclear. We previously showed that SIRT1 is highly activated in the nuclear fraction of the dentate gyrus of the chronically stressed animals and inhibits memory formation and increases anhedonic behavior during chronic stress, but specific functional targets of cytoplasmic SIRT1 are unknown. Here, we demonstrate that SIRT1 activity rapidly modulates intrinsic and synaptic properties of the dentate gyrus granule cells and anxiety behaviors through deacetylation of BK channel α subunits in control animals. Chronic stress decreases BKα channel membrane expression, and SIRT1 activity has no rapid effects on synaptic transmission or intrinsic properties in the chronically stressed animal. These results suggest SIRT1 activity rapidly modulates the physiological function of the dentate gyrus, and this modulation participates in the maladaptive stress response. Diankun Yu et al. show that deacetylase SIRT1 rapidly modulates synaptic properties of the dentate gyrus granule cells and anxiety behaviors through deacetylation of BK channel α subunits. This study provides mechanistic insight into how SIRT1 regulates fight-or-flight stress response.
Collapse
|
27
|
Storsberg S, Stryjek R, Modlińska K, Gottswinter K, D'Hanis W, Kröber A, Wernecke KE, Roskoden T, Fendt M. Predator odor induced defensive behavior in wild and laboratory rats: A comparative study. Physiol Behav 2018; 194:341-347. [DOI: 10.1016/j.physbeh.2018.06.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 06/01/2018] [Accepted: 06/08/2018] [Indexed: 01/04/2023]
|
28
|
Dunlap KD, Keane G, Ragazzi M, Lasky E, Salazar VL. Simulated predator stimuli reduce brain cell proliferation in two electric fish species, Brachyhypopomus gauderio and Apteronotus leptorhynchus. ACTA ACUST UNITED AC 2018; 220:2328-2334. [PMID: 28679791 DOI: 10.1242/jeb.158246] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 04/10/2017] [Indexed: 12/19/2022]
Abstract
The brain structure of many animals is influenced by their predators, but the cellular processes underlying this brain plasticity are not well understood. Previous studies showed that electric fish (Brachyhypopomus occidentalis) naturally exposed to high predator (Rhamdia quelen) density and tail injury had reduced brain cell proliferation compared with individuals facing few predators and those with intact tails. However, these field studies described only correlations between predator exposure and cell proliferation. Here, we used a congener Brachyhypopomus gauderio and another electric fish Apteronotus leptorhynchus to experimentally test the hypothesis that exposure to a predator stimulus and tail injury causes alterations in brain cell proliferation. To simulate predator exposure, we either amputated the tail followed by short-term (1 day) or long-term (17-18 days) recovery or repeatedly chased intact fish with a plastic rod over a 7 day period. We measured cell proliferation (PCNA+ cell density) in the telencephalon and diencephalon, and plasma cortisol, which commonly mediates stress-induced changes in brain cell proliferation. In both species, either tail amputation or simulated predator chase decreased cell proliferation in the telencephalon in a manner resembling the effect of predators in the field. In A. leptorhynchus, cell proliferation decreased drastically in the short term after tail amputation and partially rebounded after long-term recovery. In B. gauderio, tail amputation elevated cortisol levels, but repeated chasing had no effect. In A. leptorhynchus, tail amputation elevated cortisol levels in the short term but not in the long term. Thus, predator stimuli can cause reductions in brain cell proliferation, but the role of cortisol is not clear.
Collapse
Affiliation(s)
- Kent D Dunlap
- Department of Biology, Trinity College, Hartford, CT 06106, USA
| | - Geoffrey Keane
- Department of Biology, Trinity College, Hartford, CT 06106, USA
| | - Michael Ragazzi
- Department of Biology, Trinity College, Hartford, CT 06106, USA.,Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Elise Lasky
- Department of Biology, Trinity College, Hartford, CT 06106, USA
| | - Vielka L Salazar
- Department of Biology, Cape Breton University, Sydney, NS, Canada B1P 6L2
| |
Collapse
|
29
|
Shohayeb B, Diab M, Ahmed M, Ng DCH. Factors that influence adult neurogenesis as potential therapy. Transl Neurodegener 2018; 7:4. [PMID: 29484176 PMCID: PMC5822640 DOI: 10.1186/s40035-018-0109-9] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 02/16/2018] [Indexed: 12/21/2022] Open
Abstract
Adult neurogenesis involves persistent proliferative neuroprogenitor populations that reside within distinct regions of the brain. This phenomenon was first described over 50 years ago and it is now firmly established that new neurons are continually generated in distinct regions of the adult brain. The potential of enhancing the neurogenic process lies in improved brain cognition and neuronal plasticity particularly in the context of neuronal injury and neurodegenerative disorders. In addition, adult neurogenesis might also play a role in mood and affective disorders. The factors that regulate adult neurogenesis have been broadly studied. However, the underlying molecular mechanisms of regulating neurogenesis are still not fully defined. In this review, we will provide critical analysis of our current understanding of the factors and molecular mechanisms that determine neurogenesis. We will further discuss pre-clinical and clinical studies that have investigated the potential of modulating neurogenesis as therapeutic intervention in neurodegeneration.
Collapse
Affiliation(s)
- Belal Shohayeb
- 1School of Biomedical Science, Faculty of Medicine, University of Queensland, St Lucia, QLD 4067 Australia
| | - Mohamed Diab
- 2Faculty of Pharmacy, Pharos University in Alexandria, P.O. Box Sidi Gaber, Alexandria, 21311 Egypt
| | - Mazen Ahmed
- 2Faculty of Pharmacy, Pharos University in Alexandria, P.O. Box Sidi Gaber, Alexandria, 21311 Egypt
| | - Dominic Chi Hiung Ng
- 1School of Biomedical Science, Faculty of Medicine, University of Queensland, St Lucia, QLD 4067 Australia
| |
Collapse
|
30
|
Saraulli D, Costanzi M, Mastrorilli V, Farioli-Vecchioli S. The Long Run: Neuroprotective Effects of Physical Exercise on Adult Neurogenesis from Youth to Old Age. Curr Neuropharmacol 2018; 15:519-533. [PMID: 27000776 PMCID: PMC5543673 DOI: 10.2174/1570159x14666160412150223] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 03/08/2016] [Accepted: 03/16/2016] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND The rapid lengthening of life expectancy has raised the problem of providing social programs to counteract the age-related cognitive decline in a growing number of older people. Physical activity stands among the most promising interventions aimed at brain wellbeing, because of its effective neuroprotective action and low social cost. The purpose of this review is to describe the neuroprotective role exerted by physical activity in different life stages. In particular, we focus on adult neurogenesis, a process which has proved being highly responsive to physical exercise and may represent a major factor of brain health over the lifespan. METHODS The most recent literature related to the subject has been reviewed. The text has been divided into three main sections, addressing the effects of physical exercise during childhood/ adolescence, adulthood and aging, respectively. For each one, the most relevant studies, carried out on both human participants and rodent models, have been described. RESULTS The data reviewed converge in indicating that physical activity exerts a positive effect on brain functioning throughout the lifespan. However, uncertainty remains about the magnitude of the effect and its biological underpinnings. Cellular and synaptic plasticity provided by adult neurogenesis are highly probable mediators, but the mechanism for their action has yet to be conclusively established. CONCLUSION Despite alternative mechanisms of action are currently debated, age-appropriate physical activity programs may constitute a large-scale, relatively inexpensive and powerful approach to dampen the individual and social impact of age-related cognitive decline.
Collapse
Affiliation(s)
- Daniele Saraulli
- Institute of Cell Biology and Neurobiology, National Research Council, & Fondazione S. Lucia, Rome. Italy
| | - Marco Costanzi
- Department of Human Sciences, LUMSA University, Rome. Italy
| | - Valentina Mastrorilli
- Institute of Cell Biology and Neurobiology, National Research Council, & Fondazione S. Lucia, Rome. Italy
| | - Stefano Farioli-Vecchioli
- Institute of Cell Biology and Neurobiology, National Research Council, Via del Fosso di Fiorano 64, 00143 Rome. Italy
| |
Collapse
|
31
|
Schoenfeld TJ, McCausland HC, Morris HD, Padmanaban V, Cameron HA. Stress and Loss of Adult Neurogenesis Differentially Reduce Hippocampal Volume. Biol Psychiatry 2017; 82:914-923. [PMID: 28629541 PMCID: PMC5683934 DOI: 10.1016/j.biopsych.2017.05.013] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 04/18/2017] [Accepted: 05/05/2017] [Indexed: 12/09/2022]
Abstract
BACKGROUND Hippocampal volume loss is a hallmark of clinical depression. Chronic stress produces volume loss in the hippocampus in humans and atrophy of CA3 pyramidal cells and suppression of adult neurogenesis in rodents. METHODS To investigate the relationship between decreased adult neurogenesis and stress-induced changes in hippocampal structure and volume, we compared the effects of chronic unpredictable restraint stress and inhibition of neurogenesis in a rat pharmacogenetic model. RESULTS Chronic unpredictable restraint stress over 4 weeks decreased total hippocampal volume, reflecting loss of volume in all hippocampal subfields and in both dorsal and ventral hippocampus. In contrast, complete inhibition of adult neurogenesis for 4 weeks led to volume reduction only in the dentate gyrus. With prolonged inhibition of neurogenesis for 8 or 16 weeks, volume loss spread to the CA3 region, but not CA1. Combining stress and inhibition of adult neurogenesis did not have additive effects on the magnitude of volume loss but did produce a volume reduction throughout the hippocampus. One month of chronic unpredictable restraint stress and inhibition of adult neurogenesis led to atrophy of pyramidal cell apical dendrites in dorsal CA3 and to neuronal reorganization in ventral CA3. Stress also significantly affected granule cell dendrites. CONCLUSIONS The findings suggest that adult neurogenesis is required to maintain hippocampal volume but is not responsible for stress-induced volume loss.
Collapse
Affiliation(s)
- Timothy J Schoenfeld
- Section on Neuroplasticity, National Institute of Mental Health, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland.
| | - Hayley C McCausland
- Section on Neuroplasticity, National Institute of Mental Health, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - H Douglas Morris
- Nuclear Magnetic Resonance Facility, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Varun Padmanaban
- Section on Neuroplasticity, National Institute of Mental Health, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Heather A Cameron
- Section on Neuroplasticity, National Institute of Mental Health, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
32
|
Food restriction reduces neurogenesis in the avian hippocampal formation. PLoS One 2017; 12:e0189158. [PMID: 29211774 PMCID: PMC5718509 DOI: 10.1371/journal.pone.0189158] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 11/20/2017] [Indexed: 02/08/2023] Open
Abstract
The mammalian hippocampus is particularly vulnerable to chronic stress. Adult neurogenesis in the dentate gyrus is suppressed by chronic stress and by administration of glucocorticoid hormones. Post-natal and adult neurogenesis are present in the avian hippocampal formation as well, but much less is known about its sensitivity to chronic stressors. In this study, we investigate this question in a commercial bird model: the broiler breeder chicken. Commercial broiler breeders are food restricted during development to manipulate their growth curve and to avoid negative health outcomes, including obesity and poor reproductive performance. Beyond knowing that these chickens are healthier than fully-fed birds and that they have a high motivation to eat, little is known about how food restriction impacts the animals' physiology. Chickens were kept on a commercial food-restricted diet during the first 12 weeks of life, or released from this restriction by feeding them ad libitum from weeks 7–12 of life. To test the hypothesis that chronic food restriction decreases the production of new neurons (neurogenesis) in the hippocampal formation, the cell proliferation marker bromodeoxyuridine was injected one week prior to tissue collection. Corticosterone levels in blood plasma were elevated during food restriction, even though molecular markers of hypothalamic-pituitary-adrenal axis activation did not differ between the treatments. The density of new hippocampal neurons was significantly reduced in the food-restricted condition, as compared to chickens fed ad libitum, similar to findings in rats at a similar developmental stage. Food restriction did not affect hippocampal volume or the total number of neurons. These findings indicate that in birds, like in mammals, reduction in hippocampal neurogenesis is associated with chronically elevated corticosterone levels, and therefore potentially with chronic stress in general. This finding is consistent with the hypothesis that the response to stressors in the avian hippocampal formation is homologous to that of the mammalian hippocampus.
Collapse
|
33
|
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: 59] [Impact Index Per Article: 8.4] [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
|
34
|
Shen P, Hu Q, Dong M, Bai S, Liang Z, Chen Z, Li P, Hu Z, Zhong X, Zhu D, Wang H, Xie P. Venlafaxine exerts antidepressant effects possibly by activating MAPK-ERK1/2 and P13K-AKT pathways in the hippocampus. Behav Brain Res 2017; 335:63-70. [PMID: 28797602 DOI: 10.1016/j.bbr.2017.08.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/31/2017] [Accepted: 08/05/2017] [Indexed: 12/21/2022]
Abstract
Serotonin noradrenaline reuptake inhibitors are effective antidepressant drugs, which include venlafaxine and duloxetine. Venlafaxine is commonly used in a clinical context, but the molecular biological mechanisms behind its effects have not been fully determined. Here, we explored the potential biological effects of venlafaxine on mouse hippocampus. Mice were randomly divided into two groups and injected daily with 0.9% NaCl solution or venlafaxine. A GC-MS-based metabolomic approach was used to identify possible metabolic differences between these groups, and the key proteins involved in the relevant pathways were validated by western blotting. In our experiments, 27 hippocampal metabolites that distinguished the venlafaxine group from the control group were identified. These differential metabolites were subjected to Ingenuity Pathway Analysis, which revealed that they were strongly related to two metabolic pathways (MAPK-ERK1/2 and P13K-AKT signaling pathways). Six key proteins, BDNF, p-c-Raf, p-MAPK, p-MEK, p-AKT, and CREB, were verified by western blotting and the results were consistent with the differential metabolites identified by GC-MS. This study sheds light on the biological mechanisms underlying the effects of venlafaxine.
Collapse
Affiliation(s)
- Peng Shen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Qingchuan Hu
- Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China; Key Laboratory of Laboratory Medical Diagnostics of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Meixue Dong
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Shunjie Bai
- Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China; Key Laboratory of Laboratory Medical Diagnostics of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Zihong Liang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China; Department of Neurology, The Inner Mongolia Autonomous Region People's Hospital, Hohhot, Inner Mongolia, China
| | - Zhi Chen
- Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China; Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Pengfei Li
- Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Zicheng Hu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Xiaogang Zhong
- Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Dan Zhu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haiyang Wang
- Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
| | - Peng Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Neurobiology, Chongqing, China; Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China; Key Laboratory of Laboratory Medical Diagnostics of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China; Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
35
|
Homiack D, O'Cinneide E, Hajmurad S, Barrileaux B, Stanley M, Kreutz MR, Schrader LA. Predator odor evokes sex-independent stress responses in male and female Wistar rats and reduces phosphorylation of cyclic-adenosine monophosphate response element binding protein in the male, but not the female hippocampus. Hippocampus 2017; 27:1016-1029. [PMID: 28599071 DOI: 10.1002/hipo.22749] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/11/2017] [Accepted: 05/23/2017] [Indexed: 12/20/2022]
Abstract
Post-traumatic stress disorder (PTSD) is characterized by memory disturbances following trauma. Acute predator threat has emerged as an ethological model of PTSD, yet the effects of predator odor on signaling cascades associated with long-term memory remain poorly understood. In this study, we exposed male and female Wistar rats to the synthetic predator odor 2,5-dihydro-2,4,5-trimethylthiazoline (TMT) to assess behavioral and physiological responses as well as rapid modulation of signal transduction cascades associated with learning and memory in the male and female hippocampus. During exposure to TMT in the homecage, both male and female animals displayed robust immobility, avoidance, and altered activity as a function of time. Physiologically, TMT exposure increased circulating corticosterone and blood glucose in both male and female rodents, suggesting that TMT evokes sex-independent behavioral and physiological responses. With respect to signal transduction, TMT exposure rapidly reduced phosphorylation of cyclic-adenosine monophosphate response element binding protein (CREB) in the male, but not the female hippocampus. Furthermore, TMT exposure reduced phosphorylation of extracellular signal-regulated kinase 1/2 and increased nuclear expression of the synapto-nuclear messenger protein Jacob in the male hippocampus, consistent with activation of the CREB shut-off pathway. In a follow-up behavioral experiment, post-training exposure to TMT did not affect spatial water maze performance of male rats. However, male rats re-introduced to the context in which TMT had previously been presented displayed avoidance and hyperactivity, but not freezing behavior or elevated corticosterone responses, suggesting that TMT exposure supports a form of contextual conditioning which is not characterized by immobility. Taken together, our findings suggest that TMT evokes similar behavioral and physiological responses in male and female Wistar rats, but affects distinct signaling cascades in the male and female hippocampus which may contribute to behavioral disruptions associated with predator exposure.
Collapse
Affiliation(s)
- Damek Homiack
- Neuroscience Program, Brain Institute, Tulane University, New Orleans, Louisiana, 70118
| | - Emma O'Cinneide
- Neuroscience Program, Brain Institute, Tulane University, New Orleans, Louisiana, 70118
| | - Sema Hajmurad
- Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, 70118
| | - Brett Barrileaux
- Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, 70118
| | - Mary Stanley
- Neuroscience Program, Brain Institute, Tulane University, New Orleans, Louisiana, 70118.,Infectious Disease and Microbiome Program, Broad Institute, Cambridge, Massachusetts, 02142
| | - Michael R Kreutz
- RG Neuroplasticity, Leibniz-Institute for Neurobiology, Magdeburg, Germany.,Leibniz Group 'Dendritic Organelles and Synaptic Function', Hamburg, Germany
| | - Laura A Schrader
- Neuroscience Program, Brain Institute, Tulane University, New Orleans, Louisiana, 70118.,Cell and Molecular Biology, Tulane University, New Orleans, Louisiana, 70118
| |
Collapse
|
36
|
Vignisse J, Sambon M, Gorlova A, Pavlov D, Caron N, Malgrange B, Shevtsova E, Svistunov A, Anthony DC, Markova N, Bazhenova N, Coumans B, Lakaye B, Wins P, Strekalova T, Bettendorff L. Thiamine and benfotiamine prevent stress-induced suppression of hippocampal neurogenesis in mice exposed to predation without affecting brain thiamine diphosphate levels. Mol Cell Neurosci 2017; 82:126-136. [PMID: 28506637 DOI: 10.1016/j.mcn.2017.05.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 05/09/2017] [Accepted: 05/12/2017] [Indexed: 12/31/2022] Open
Abstract
Thiamine is essential for normal brain function and its deficiency causes metabolic impairment, specific lesions, oxidative damage and reduced adult hippocampal neurogenesis (AHN). Thiamine precursors with increased bioavailability, especially benfotiamine, exert neuroprotective effects not only for thiamine deficiency (TD), but also in mouse models of neurodegeneration. As it is known that AHN is impaired by stress in rodents, we exposed C57BL6/J mice to predator stress for 5 consecutive nights and studied the proliferation (number of Ki67-positive cells) and survival (number of BrdU-positive cells) of newborn immature neurons in the subgranular zone of the dentate gyrus. In stressed mice, the number of Ki67- and BrdU-positive cells was reduced compared to non-stressed animals. This reduction was prevented when the mice were treated (200mg/kg/day in drinking water for 20days) with thiamine or benfotiamine, that were recently found to prevent stress-induced behavioral changes and glycogen synthase kinase-3β (GSK-3β) upregulation in the CNS. Moreover, we show that thiamine and benfotiamine counteract stress-induced bodyweight loss and suppress stress-induced anxiety-like behavior. Both treatments induced a modest increase in the brain content of free thiamine while the level of thiamine diphosphate (ThDP) remained unchanged, suggesting that the beneficial effects observed are not linked to the role of this coenzyme in energy metabolism. Predator stress increased hippocampal protein carbonylation, an indicator of oxidative stress. This effect was antagonized by both thiamine and benfotiamine. Moreover, using cultured mouse neuroblastoma cells, we show that in particular benfotiamine protects against paraquat-induced oxidative stress. We therefore hypothesize that thiamine compounds may act by boosting anti-oxidant cellular defenses, by a mechanism that still remains to be unveiled. Our study demonstrates, for the first time, that thiamine and benfotiamine prevent stress-induced inhibition of hippocampal neurogenesis and accompanying physiological changes. The present data suggest that thiamine precursors with high bioavailability might be useful as a complementary therapy in several neuropsychiatric disorders.
Collapse
Affiliation(s)
| | | | - Anna Gorlova
- Laboratory of Psychiatric Neurobiology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Dmitrii Pavlov
- Laboratory of Psychiatric Neurobiology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Nicolas Caron
- GIGA-Neurosciences, University of Liege, Liege, Belgium
| | | | - Elena Shevtsova
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Moscow, Russia
| | - Andrey Svistunov
- Laboratory of Psychiatric Neurobiology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Natalyia Markova
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Moscow, Russia; Department of Pharmacology, Oxford University, Oxford, UK; Institute of General Pathology and Pathophysiology, Moscow 125 315, Russia; Department of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | - Natalyia Bazhenova
- Laboratory of Psychiatric Neurobiology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; Institute of General Pathology and Pathophysiology, Moscow 125 315, Russia; Department of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands
| | | | | | - Pierre Wins
- GIGA-Neurosciences, University of Liege, Liege, Belgium
| | - Tatyana Strekalova
- Laboratory of Psychiatric Neurobiology, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; Department of Neuroscience, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands.
| | | |
Collapse
|
37
|
Tzeng WY, Wu HH, Wang CY, Chen JC, Yu L, Cherng CG. Sex Differences in Stress and Group Housing Effects on the Number of Newly Proliferated Cells and Neuroblasts in Middle-Aged Dentate Gyrus. Front Behav Neurosci 2017; 10:249. [PMID: 28119581 PMCID: PMC5220061 DOI: 10.3389/fnbeh.2016.00249] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2016] [Accepted: 12/19/2016] [Indexed: 11/13/2022] Open
Abstract
Sex differences in stress and coping responses have been frequently documented in aged people, while whether such differences in aged people may appear at the middle age are unknown. This study was undertaken to study the impact of acute stress and social interaction on early neurogenesis in the dentate gyrus (DG) and hippocampus-related memory in two sexes of middle-aged mice. The number of newly proliferated cells, neuroblasts in DG, the object recognition and location memory in 9-month-old male and female C57BL/6N mice were assessed under baseline conditions as well as following an acute stressor regimen and group housing. Three conspecific companions, serving as "the housing group," were used to model the social interaction throughout the stressor regimen. Males had lower numbers of newly proliferated cells and neuroblasts under baseline conditions as compared to females. The stressor regimen caused rapid decreases in the number of newly proliferated cells and neuroblasts in female DG but no obvious changes were observed in male DG. Group housing, regardless of companions' age, prevented the stress-induced decreases in the number of newly proliferated cells and neuroblasts in female DG. In contrast, the presence of young or age-matched companions potentiated the stress effect in males by decreasing the number of newly proliferated cells and neuroblasts. Finally, neither the stressor regimen nor group housing affected mouse performances in the object recognition and location memory in either sex. These findings, taken together, provide evidence to support a notion that middle-aged females appear to demonstrate more stress susceptibility on early neurogenesis in DG as compared to middle-aged males, although the hippocampus-related memory performances are comparable and not affected by stress in these males and females. Experiencing stress, middle-aged females are more prone to benefit from social interaction as compared to middle-aged males in this regard. We suggest, accordingly, that involving social interaction may afford a therapeutic advance in preventing stress-produced decreases in early neurogenesis in middle-aged females' DG.
Collapse
Affiliation(s)
- Wen-Yu Tzeng
- Department of Physiology, National Cheng Kung University College of Medicine Tainan, Taiwan
| | - Hsin-Hua Wu
- Department of Physiology, National Cheng Kung University College of Medicine Tainan, Taiwan
| | - Ching-Yi Wang
- Institute of Basic Medical Sciences, National Cheng Kung University College of Medicine Tainan, Taiwan
| | - Jin-Chung Chen
- Graduate Institute of Biomedical Sciences, Chang Gung University Taoyuan, Taiwan
| | - Lung Yu
- Department of Physiology, National Cheng Kung University College of MedicineTainan, Taiwan; Institute of Basic Medical Sciences, National Cheng Kung University College of MedicineTainan, Taiwan
| | - Chianfang G Cherng
- Department of Health Psychology, Chang Jung Christian University Tainan, Taiwan
| |
Collapse
|
38
|
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: 1.6] [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]
|
39
|
Fokos S, Pavlidis M, Yiotis T, Tsalafouta A, Papandroulakis N, Dermon CR. Early life low intensity stress experience modifies acute stress effects on juvenile brain cell proliferation of European sea bass (D. Labrax). Behav Brain Res 2016; 317:109-121. [PMID: 27638037 DOI: 10.1016/j.bbr.2016.09.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 09/04/2016] [Accepted: 09/11/2016] [Indexed: 02/06/2023]
Abstract
Early life adversity may be critical for the brain structural plasticity that in turn would influence juvenile behaviour. To address this, we questioned whether early life environment has an impact on stress responses latter in life, using European sea bass, Dicentrarchus labrax, as a model organism. Unpredictable chronic low intensity stress (UCLIS), using a variety of moderate intensity stressors, was applied during two early ontogenetic stages, flexion or formation all fins. At juvenile stage, fish were exposed to acute stress and plasma cortisol, brain mRNA expression of corticosteroid receptors' genes (gr1, gr2, mr) and brain cell proliferation (using BrdU immunohistochemistry) were determined in experimental and matched controls. UCLIS treatment specifically decreased brain gr1 expression in juveniles, but had no effect on the juvenile brain cell proliferation pattern within the major neurogenic zones studied of dorsal (Dm, Dld) and ventral (Vv) telencephalic, preoptic (NPO) areas, periventricular tectum gray zone (PGZ) and valvula cerebellum (VCe). In contrast, exposure to acute stress induced significant plasma cortisol rise, decreases of cerebral cell proliferation in juveniles, not previously exposed to UCLIS, but no effect detected on the expression levels of gr1, gr2 and mr in all groups of different early life history. Interestingly, juveniles with UCLIS history showed modified responses to acute stress, attenuating acute stress-induced cell proliferation decreases, indicating a long-lasting effect of early life treatment. Taken together, early life mild stress experience influences an acute stress plasticity end-point, cerebral cell proliferation, independently of the stress-axis activation, possibly leading to more effective coping styles.
Collapse
Affiliation(s)
- S Fokos
- Dept. of Biology, Human and Animal Physiology Lab, University of Patras, Greece(1)
| | - M Pavlidis
- Dept. of Biology, University of Crete, Greece
| | - T Yiotis
- Dept. of Biology, Human and Animal Physiology Lab, University of Patras, Greece(1)
| | - A Tsalafouta
- Dept. of Biology, University of Crete, Greece; Aquaculture Institute, Hellenic Centre Marine Research, Crete, Greece
| | - N Papandroulakis
- Aquaculture Institute, Hellenic Centre Marine Research, Crete, Greece
| | - C R Dermon
- Dept. of Biology, Human and Animal Physiology Lab, University of Patras, Greece(1).
| |
Collapse
|
40
|
Lieberwirth C, Pan Y, Liu Y, Zhang Z, Wang Z. Hippocampal adult neurogenesis: Its regulation and potential role in spatial learning and memory. Brain Res 2016; 1644:127-40. [PMID: 27174001 PMCID: PMC5064285 DOI: 10.1016/j.brainres.2016.05.015] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 05/05/2016] [Accepted: 05/08/2016] [Indexed: 12/24/2022]
Abstract
Adult neurogenesis, defined here as progenitor cell division generating functionally integrated neurons in the adult brain, occurs within the hippocampus of numerous mammalian species including humans. The present review details various endogenous (e.g., neurotransmitters) and environmental (e.g., physical exercise) factors that have been shown to influence hippocampal adult neurogenesis. In addition, the potential involvement of adult-generated neurons in naturally-occurring spatial learning behavior is discussed by summarizing the literature focusing on traditional animal models (e.g., rats and mice), non-traditional animal models (e.g., tree shrews), as well as natural populations (e.g., chickadees and Siberian chipmunk).
Collapse
Affiliation(s)
| | - Yongliang Pan
- Program in Molecular and Translational Medicine, School of Medicine, Huzhou University, Huzhou 313000, PR China; State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang District, Beijing 100101, PR China.
| | - Yan Liu
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306-1270, USA
| | - Zhibin Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents in Agriculture, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang District, Beijing 100101, PR China
| | - Zuoxin Wang
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL 32306-1270, USA
| |
Collapse
|
41
|
Trading new neurons for status: Adult hippocampal neurogenesis in eusocial Damaraland mole-rats. Neuroscience 2016; 324:227-37. [DOI: 10.1016/j.neuroscience.2016.03.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 02/19/2016] [Accepted: 03/07/2016] [Indexed: 11/21/2022]
|
42
|
Anxiety- and Depression-Like States Lead to Pronounced Olfactory Deficits and Impaired Adult Neurogenesis in Mice. J Neurosci 2016; 36:518-31. [PMID: 26758842 DOI: 10.1523/jneurosci.2817-15.2016] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Numerous clinical reports underscore the frequency of olfactory impairments in patients suffering from major depressive disorders (MDDs), yet the underlying physiopathological mechanisms remain poorly understood. We hypothesized that one key link between olfactory deficits and MDD lies in hypercortisolemia, a cardinal symptom of MDD. Corticosterone (CORT) is known to negatively correlate with hippocampal neurogenesis, yet its effects on olfactory neurogenesis and olfaction remain unknown. Here we used a rodent model of anxiety/depression-like states, which is based on chronic CORT administration and studied the effects of the antidepressant fluoxetine (FLX) on behavior, olfaction, and adult neurogenesis in the dentate gyrus (DG), olfactory bulb (OB), and the olfactory epithelium (OE). Chronic CORT had no effect on cell proliferation in the OE or on olfactory sensory neurons projecting to the OB, but induced pronounced deficits in olfactory acuity, fine discrimination of odorants and olfactory memory. These alterations were accompanied by a significant decrease in the number of adult-born neurons in both the DG and OB. Remarkably, FLX not only reversed depression-like states as expected, but also improved olfactory acuity, memory, and restored impaired adult neurogenesis. However, fine olfactory discrimination was not restored. Morphological analysis of adult-born neurons in both the DG and the OB showed that dendritic complexity was not significantly affected by CORT, but was increased by FLX. These findings demonstrate an essential role for glucocorticoids in triggering olfactory impairments in MDD and highlight a novel therapeutic effect of FLX. SIGNIFICANCE STATEMENT Increasing clinical reports show that major depression is characterized by pronounced olfactory deficits, yet the underlying mechanisms remain unknown. In this work, we used an endocrine model of depression to study whether hypothalamic-pituitary-adrenal axis perturbation could be sufficient to provoke olfactory impairments. We found that chronic corticosterone not only induces marked deficits in olfactory acuity, fine discrimination and olfactory memory, but also significantly decreases bulbar and hippocampal neurogenesis. Importantly, the antidepressant fluoxetine restores both adult neurogenesis and depressive states, and improves most olfactory functions. Our data reveal that impairment of hypothalamic-pituitary-adrenal axis during depression can lead to olfactory deficits and that the neurogenic effects of selective serotonin reuptake inhibitor antidepressants can successfully restore certain olfactory functions.
Collapse
|
43
|
Wulsin AC, Solomon MB, Privitera MD, Danzer SC, Herman JP. Hypothalamic-pituitary-adrenocortical axis dysfunction in epilepsy. Physiol Behav 2016; 166:22-31. [PMID: 27195458 DOI: 10.1016/j.physbeh.2016.05.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 04/04/2016] [Accepted: 05/11/2016] [Indexed: 12/27/2022]
Abstract
Epilepsy is a common neurological disease, affecting 2.4million people in the US. Among the many different forms of the disease, temporal lobe epilepsy (TLE) is one of the most frequent in adults. Recent studies indicate the presence of a hyperactive hypothalamopituitary- adrenocortical (HPA) axis and elevated levels of glucocorticoids in TLE patients. Moreover, in these patients, stress is a commonly reported trigger of seizures, and stress-related psychopathologies, including depression and anxiety, are highly prevalent. Elevated glucocorticoids have been implicated in the development of stress-related psychopathologies. Similarly, excess glucocorticoids have been found to increase neuronal excitability, epileptiform activity and seizure susceptibility. Thus, patients with TLE may generate abnormal stress responses that both facilitate ictal discharges and increase vulnerability for the development of comorbid psychopathologies. Here, we will examine the evidence that the HPA axis is disrupted in TLE, consider potential mechanisms by which this might occur, and discuss the implications of HPA dysfunction for seizuretriggering and psychiatric comorbidities.
Collapse
Affiliation(s)
- Aynara C Wulsin
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, College of Medicine, Cincinnati, OH, United States; Neuroscience Program, University of Cincinnati, College of Medicine, Cincinnati, OH, United States; Department of Anesthesia, Cincinnati Childrens Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, Cincinnati Childrens Hospital Medical Center, Cincinnati, OH, United States.
| | - Matia B Solomon
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, College of Medicine, Cincinnati, OH, United States; Neuroscience Program, University of Cincinnati, College of Medicine, Cincinnati, OH, United States
| | - Michael D Privitera
- Department of Neurology, Neuroscience Institute, University of Cincinnati, Cincinnati, OH, United States
| | - Steve C Danzer
- Neuroscience Program, University of Cincinnati, College of Medicine, Cincinnati, OH, United States; Department of Anesthesia, Cincinnati Childrens Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, Cincinnati Childrens Hospital Medical Center, Cincinnati, OH, United States
| | - James P Herman
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, College of Medicine, Cincinnati, OH, United States; Neuroscience Program, University of Cincinnati, College of Medicine, Cincinnati, OH, United States.
| |
Collapse
|
44
|
The effects of hormones and physical exercise on hippocampal structural plasticity. Front Neuroendocrinol 2016; 41:23-43. [PMID: 26989000 DOI: 10.1016/j.yfrne.2016.03.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 03/02/2016] [Accepted: 03/08/2016] [Indexed: 01/22/2023]
Abstract
The hippocampus plays an integral role in certain aspects of cognition. Hippocampal structural plasticity and in particular adult hippocampal neurogenesis can be influenced by several intrinsic and extrinsic factors. Here we review how hormones (i.e., intrinsic modulators) and physical exercise (i.e., an extrinsic modulator) can differentially modulate hippocampal plasticity in general and adult hippocampal neurogenesis in particular. Specifically, we provide an overview of the effects of sex hormones, stress hormones, and metabolic hormones on hippocampal structural plasticity and adult hippocampal neurogenesis. In addition, we also discuss how physical exercise modulates these forms of hippocampal plasticity, giving particular emphasis on how this modulation can be affected by variables such as exercise regime, duration, and intensity. Understanding the neurobiological mechanisms underlying the modulation of hippocampal structural plasticity by intrinsic and extrinsic factors will impact the design of new therapeutic approaches aimed at restoring hippocampal plasticity following brain injury or neurodegeneration.
Collapse
|
45
|
Korn MJ, Mandle QJ, Parent JM. Conditional Disabled-1 Deletion in Mice Alters Hippocampal Neurogenesis and Reduces Seizure Threshold. Front Neurosci 2016; 10:63. [PMID: 26941603 PMCID: PMC4766299 DOI: 10.3389/fnins.2016.00063] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/10/2016] [Indexed: 11/13/2022] Open
Abstract
Many animal models of temporal lobe epilepsy (TLE) exhibit altered neurogenesis arising from progenitors within the dentate gyrus subgranular zone (SGZ). Aberrant integration of new neurons into the existing circuit is thought to contribute to epileptogenesis. In particular, adult-born neurons that exhibit ectopic migration and hilar basal dendrites (HBDs) are suggested to be pro-epileptogenic. Loss of reelin signaling may contribute to these morphological changes in patients with epilepsy. We previously demonstrated that conditional deletion of the reelin adaptor protein, disabled-1 (Dab1), from postnatal mouse SGZ progenitors generated dentate granule cells (DGCs) with abnormal dendritic development and ectopic placement. To determine whether the early postnatal loss of reelin signaling is epileptogenic, we conditionally deleted Dab1 in neural progenitors and their progeny on postnatal days 7–8 and performed chronic video-EEG recordings 8–10 weeks later. Dab1-deficient mice did not have spontaneous seizures but exhibited interictal epileptiform abnormalities and a significantly reduced latency to pilocarpine-induced status epilepticus. After chemoconvulsant treatment, over 90% of mice deficient for Dab1 developed generalized motor convulsions with tonic-clonic movements, rearing, and falling compared to <20% of wild-type mice. Recombination efficiency, measured by Cre reporter expression, inversely correlated with time to the first sustained seizure. These pro-epileptogenic changes were associated with decreased neurogenesis and increased numbers of hilar ectopic DGCs. Interestingly, neurons co-expressing the Cre reporter comprised a fraction of these hilar ectopic DGCs cells, suggesting a non-cell autonomous effect for the loss of reelin signaling. We also noted a dispersion of the CA1 pyramidal layer, likely due to hypomorphic effects of the conditional Dab1 allele, but this abnormality did not correlate with seizure susceptibility. These findings suggest that the misplacement or reduction of postnatally-generated DGCs contributes to aberrant circuit development and hyperexcitability, but aberrant neurogenesis after conditional Dab1 deletion alone is not sufficient to produce spontaneous seizures.
Collapse
Affiliation(s)
- Matthew J Korn
- Department of Neurology, University of Michigan Medical Center Ann Arbor, MI, USA
| | - Quinton J Mandle
- Department of Neurology, University of Michigan Medical Center Ann Arbor, MI, USA
| | - Jack M Parent
- Department of Neurology, University of Michigan Medical CenterAnn Arbor, MI, USA; VA Ann Arbor Healthcare SystemAnn Arbor, MI, USA
| |
Collapse
|
46
|
Dunlap KD, Tran A, Ragazzi MA, Krahe R, Salazar VL. Predators inhibit brain cell proliferation in natural populations of electric fish, Brachyhypopomus occidentalis. Proc Biol Sci 2016; 283:20152113. [PMID: 26842566 PMCID: PMC4760157 DOI: 10.1098/rspb.2015.2113] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 01/08/2016] [Indexed: 11/12/2022] Open
Abstract
Compared with laboratory environments, complex natural environments promote brain cell proliferation and neurogenesis. Predators are one important feature of many natural environments, but, in the laboratory, predatory stimuli tend to inhibit brain cell proliferation. Often, laboratory predatory stimuli also elevate plasma glucocorticoids, which can then reduce brain cell proliferation. However, it is unknown how natural predators affect cell proliferation or whether glucocorticoids mediate the neurogenic response to natural predators. We examined brain cell proliferation in six populations of the electric fish, Brachyhypopomus occidentalis, exposed to three forms of predator stimuli: (i) natural variation in the density of predatory catfish; (ii) tail injury, presumably from predation attempts; and (iii) the acute stress of capture. Populations with higher predation pressure had lower density of proliferating (PCNA+) cells, and fish with injured tails had lower proliferating cell density than those with intact tails. However, plasma cortisol did not vary at the population level according to predation pressure or at the individual level according to tail injury. Capture stress significantly increased cortisol, but only marginally decreased cell proliferation. Thus, it appears that the presence of natural predators inhibits brain cell proliferation, but not via mechanisms that depend on changes in basal cortisol levels. This study is the first demonstration of predator-induced alteration of brain cell proliferation in a free-living vertebrate.
Collapse
Affiliation(s)
- Kent D Dunlap
- Department of Biology, Trinity College, Hartford, CT 06106, USA
| | - Alex Tran
- Department of Biology, McGill University, Montreal, Quebec, Canada H3A 1B1
| | | | - Rüdiger Krahe
- Department of Biology, McGill University, Montreal, Quebec, Canada H3A 1B1
| | - Vielka L Salazar
- Department of Biology, Cape Breton University, Sydney, Nova Scotia, Canada B1P 6L2
| |
Collapse
|
47
|
Rizi AA, Reisi P, Naghsh N. Effect of forced treadmill exercise and blocking of opioid receptors with naloxone on memory in male rats. Adv Biomed Res 2016; 5:20. [PMID: 26962522 PMCID: PMC4770631 DOI: 10.4103/2277-9175.175906] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 09/08/2015] [Indexed: 12/16/2022] Open
Abstract
Background: The forced treadmill running can influence the opioid contents of the brain, through both effects of exercise and the effects of stress caused by coercion. Since opioids can cause negative effects on brain functions, this study aimed to evaluate the effect of forced treadmill exercise and blocking of opioid receptors with naloxone on memory in male rats. Materials and Methods: Experimental groups were the control, the exercise, the naloxone, and the naloxone exercise. The exercise program was treadmill running at 22 m/min at 0° inclination for 50 min/day, 6 days/week, for 4 weeks. Naloxone (1 mg/kg) was injected 5 min before the treadmill running. Morris water maze and passive avoidance learning tests were used for evaluation of memory. Acquisition phase of both tests was performed before interventions, and memory was evaluated 1-day and 1-week after the last session of exercise and treatments. Results: Our data showed that forced exercise impaired performance in passive avoidance learning test (P < 0.05 and P <0.01, 1-day, and 1-week after the last session of exercise and treatments, respectively). Spatial memory was only impaired after 1-week in the exercise group. Naloxone had no significant effect on memory in the control group. However, it improved memory in the exercise group, as there was no significant difference between the control and the naloxone exercise in both tests. Conclusion: The data correspond to the possibility that opioidergic system may have mediatory roles in exercise-induced responses in forced exercise. These roles are likely harmful for memory.
Collapse
Affiliation(s)
- Atefeh Asadi Rizi
- Department of Biology, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
| | - Parham Reisi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Applied Physiology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran; Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nooshin Naghsh
- Department of Biology, Falavarjan Branch, Islamic Azad University, Isfahan, Iran
| |
Collapse
|
48
|
Motta-Teixeira LC, Takada SH, Machado-Nils AV, Nogueira MI, Xavier GF. Spatial learning and neurogenesis: Effects of cessation of wheel running and survival of novel neurons by engagement in cognitive tasks. Hippocampus 2016; 26:794-803. [PMID: 26669934 DOI: 10.1002/hipo.22560] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2015] [Indexed: 01/01/2023]
Abstract
Physical exercise stimulates cell proliferation in the adult dentate gyrus and facilitates acquisition and/or retention of hippocampal-dependent tasks. It is established that regular physical exercise improves cognitive performance. However, it is unclear for how long these benefits last after its interruption. Independent groups of rats received both free access to either unlocked (EXE Treatment) or locked (No-EXE Treatment) running wheels for 7 days, and daily injections of bromodeoxyuridine (BrdU) in the last 3 days. After a time delay period of either 1, 3, or 6 weeks without training, the animals were tested in the Morris water maze (MWM) either in a working memory task dependent on hippocampal function (MWM-HD) or in a visible platform searching task, independent on hippocampal function (MWM-NH). Data confirmed that exposure of rats to 7 days of spontaneous wheel running increases cell proliferation and neurogenesis. In contrast, neurogenesis was not accompanied by significant improvements of performance in the working memory version of the MWM. Longer time delays between the end of exercise and the beginning of cognitive training in the MWM resulted in lower cell survival; that is, the number of novel surviving mature neurons was decreased when this delay was 6 weeks as compared with when it was 1 week. In addition, data showed that while exposure to the MWM-HD working memory task substantially increased survival of novel neurons, exposure to the MWM-NH task did not, thus indicating that survival of novel dentate gyrus neurons depends on the engagement of this brain region in performance of cognitive tasks. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lívia Clemente Motta-Teixeira
- Department of Physiology, Institute of Biosciences, University of São Paulo, Rua Do Matão, Travessa 14, N. 101, São Paulo, 05508-090, SP, Brazil
| | - Silvia Honda Takada
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, Av. Professor Lineu Prestes, 2415, São Paulo, 05508-000, SP, Brazil
| | - Aline Vilar Machado-Nils
- Department of Physiology, Institute of Biosciences, University of São Paulo, Rua Do Matão, Travessa 14, N. 101, São Paulo, 05508-090, SP, Brazil
| | - Maria Inês Nogueira
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, Av. Professor Lineu Prestes, 2415, São Paulo, 05508-000, SP, Brazil
| | - Gilberto Fernando Xavier
- Department of Physiology, Institute of Biosciences, University of São Paulo, Rua Do Matão, Travessa 14, N. 101, São Paulo, 05508-090, SP, Brazil
| |
Collapse
|
49
|
Gesmundo I, Villanova T, Gargantini E, Arvat E, Ghigo E, Granata R. The Mineralocorticoid Agonist Fludrocortisone Promotes Survival and Proliferation of Adult Hippocampal Progenitors. Front Endocrinol (Lausanne) 2016; 7:66. [PMID: 27379018 PMCID: PMC4910464 DOI: 10.3389/fendo.2016.00066] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 06/01/2016] [Indexed: 11/13/2022] Open
Abstract
Glucocorticoid receptor (GR) activation has been shown to reduce adult hippocampal progenitor cell proliferation and neurogenesis. By contrast, mineralocorticoid receptor (MR) signaling is associated with neuronal survival in the dentate gyrus of the hippocampus, and impairment of hippocampal MR has been linked to pathological conditions, such as depression or neurodegenerative disorders. Here, we aimed to further clarify the protective role of MR in adult hippocampal neurons by studying the survival and proliferative effects of the highly potent MR agonist fludrocortisone (Fludro) in adult rat hippocampal progenitor cells (AHPs), along with the associated signaling mechanisms. Fludro, which upregulated MR but not GR expression, increased survival and proliferation and prevented apoptosis in AHPs cultured in growth factor-deprived medium. These effects were blunted by the MR antagonist spironolactone and by high doses of the GR agonist dexamethasone. Moreover, they involved signaling through cAMP/protein kinase A (PKA)/cAMP response element-binding protein, phosphoinositide 3-kinase (PI3K)/Akt and its downstream targets glycogen synthase kinase-3β (GSK-3β) and mammalian target of rapamycin. Furthermore, Fludro attenuated the detrimental effects of amyloid-β peptide 1-42 (Aβ1-42) on cell survival, proliferation, and apoptosis in AHPs, and increased the phosphorylation of both PI3K/Akt and GSK-3β, which was reduced by Aβ1-42. Finally, Fludro blocked Aβ1-42-induced hyperphosphorylation of Tau protein, which is a main feature of Alzheimer's disease. Overall, these results are the first to show the protective and proliferative role of Fludro in AHPs, suggesting the potential therapeutic importance of targeting MR for increasing hippocampal neurogenesis and for treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Iacopo Gesmundo
- Laboratory of Molecular and Cellular Endocrinology, Department of Medical Sciences, University of Torino, Torino, Italy
- Department of Medical Sciences, Division of Endocrinology, Diabetes and Metabolism, University of Torino, Torino, Italy
| | - Tania Villanova
- Laboratory of Molecular and Cellular Endocrinology, Department of Medical Sciences, University of Torino, Torino, Italy
- Department of Medical Sciences, Division of Endocrinology, Diabetes and Metabolism, University of Torino, Torino, Italy
| | - Eleonora Gargantini
- Laboratory of Molecular and Cellular Endocrinology, Department of Medical Sciences, University of Torino, Torino, Italy
- Department of Medical Sciences, Division of Endocrinology, Diabetes and Metabolism, University of Torino, Torino, Italy
| | - Emanuela Arvat
- Department of Medical Sciences, Division of Oncological Endocrinology, University of Torino, Torino, Italy
| | - Ezio Ghigo
- Department of Medical Sciences, Division of Endocrinology, Diabetes and Metabolism, University of Torino, Torino, Italy
| | - Riccarda Granata
- Laboratory of Molecular and Cellular Endocrinology, Department of Medical Sciences, University of Torino, Torino, Italy
- Department of Medical Sciences, Division of Endocrinology, Diabetes and Metabolism, University of Torino, Torino, Italy
- *Correspondence: Riccarda Granata,
| |
Collapse
|
50
|
|