151
|
Davis EG, Keller J, Hallmayer J, Pankow HR, Murphy GM, Gotlib IH, Schatzberg AF. Corticotropin-releasing factor 1 receptor haplotype and cognitive features of major depression. Transl Psychiatry 2018; 8:5. [PMID: 29317606 PMCID: PMC5802461 DOI: 10.1038/s41398-017-0051-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 09/13/2017] [Indexed: 12/22/2022] Open
Abstract
Corticotropin-releasing factor signaling through CRF receptor type 1 (CRF1) has been shown to contribute to learning and memory function. A haplotype of alleles T-A-T in a set of common polymorphisms in the gene encoding for CRF1 (CRHR1) has been associated with both depression vulnerability and alterations in cognitive functioning. The present study investigated the relations between the TAT haplotype and specific symptoms of depression, self-reported ruminative behaviors, and neuropsychological performance on a learning and memory task. Participants were adults with major depression with and without psychotic features (N = 406). Associations were examined between TAT haplotype and endorsement of depression symptoms from diagnostic interviews, scores on the rumination response scale (RRS), and verbal memory performance on the California Verbal Learning Test-II (CVLT-II). All analyses included depression subtype, age, and sex as covariates; CVLT-II analyses also included evening cortisol levels. Across the entire sample, carriers of more copies of the TAT haplotype reported greater endorsement of the symptom describing difficulty concentrating and making decisions. In separate subsamples, TAT homozygotes had higher rumination scores on the RRS, both brooding and reflection subscales, and more TAT copies were associated with poorer CVLT-II performance in both total learning and free recall trials. These data demonstrate that the CRHR1 TAT haplotype is associated with cognitive features of depression including difficulty with decision-making, higher rumination, and poorer learning and memory. It will be important in future research to identify the specific molecular mechanisms for CRF1 signaling that contribute to depression-related cognitive dysfunction.
Collapse
Affiliation(s)
- Elena Goetz Davis
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, USA.
| | - Jennifer Keller
- 0000000419368956grid.168010.eDepartment of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, USA
| | - Joachim Hallmayer
- 0000000419368956grid.168010.eDepartment of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, USA
| | - Heather Ryan Pankow
- 0000000419368956grid.168010.eDepartment of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, USA
| | - Greer M. Murphy
- 0000000419368956grid.168010.eDepartment of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, USA
| | - Ian H. Gotlib
- 0000000419368956grid.168010.eDepartment of Psychology, Stanford University, Stanford, USA
| | - Alan F. Schatzberg
- 0000000419368956grid.168010.eDepartment of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, USA
| |
Collapse
|
152
|
de Andrade JS, Céspedes IC, Abrão RO, da Silva JM, Ceneviva R, Ribeiro DA, Bittencourt JC, Viana MB. Effects of acute restraint and unpredictable chronic mild stress on brain corticotrophin releasing factor mRNA in the elevated T-maze. Behav Brain Res 2018; 337:139-150. [DOI: 10.1016/j.bbr.2017.09.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/15/2017] [Accepted: 09/17/2017] [Indexed: 11/29/2022]
|
153
|
Sadeghi M, Reisi P, Radahmadi M. The effects of CCK-8S on spatial memory and long-term potentiation at CA1 during induction of stress in rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2017; 20:1368-1376. [PMID: 29238473 PMCID: PMC5722998 DOI: 10.22038/ijbms.2017.9619] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Objectives Cholecystokinin (CCK) has been proposed as a mediator in stress. However, it is still not fully documented what are its effects. We aimed to evaluate the effects of systemic administration of CCK exactly before induction of stress on spatial memory and synaptic plasticity at CA1 in rats. Materials and Methods Male Wistar rats were divided into 4 groups: the control, the control-CCK, the stress and the stress-CCK. Restraint stress was induced 6 hr per day, for 24 days. Cholecystokinin sulfated octapeptide (CCK-8S) was injected (1.6 µg/kg, IP) before each session of stress induction. Spatial memory was evaluated by Morris water maze test. Long-term potentiation (LTP) in Schaffer collateral-CA1 synapses was assessed (by 100 Hz tetanization) in order to investigate synaptic plasticity. Results Stress impaired spatial memory significantly (P<0.01). CCK in the control rats improved memory (P<0.05), and prevented the impairments in the stress group. With respect to the control group, both fEPSP amplitude and slope were significantly (P<0.05) decreased in the stress group. However, there were no differences between responses of the control-CCK and Stress-CCK groups compared to the control group. Conclusion The present results suggest that high levels of CCK-8S during induction of stress can modulate the destructive effects of stress on hippocampal synaptic plasticity and memory. Therefore, the mediatory effects of CCK in stress are likely as compensatory responses.
Collapse
Affiliation(s)
- Malihe Sadeghi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parham Reisi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Radahmadi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
154
|
Spencer SJ, Korosi A, Layé S, Shukitt-Hale B, Barrientos RM. Food for thought: how nutrition impacts cognition and emotion. NPJ Sci Food 2017; 1:7. [PMID: 31304249 PMCID: PMC6550267 DOI: 10.1038/s41538-017-0008-y] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 07/24/2017] [Accepted: 08/10/2017] [Indexed: 01/27/2023] Open
Abstract
More than one-third of American adults are obese and statistics are similar worldwide. Caloric intake and diet composition have large and lasting effects on cognition and emotion, especially during critical periods in development, but the neural mechanisms for these effects are not well understood. A clear understanding of the cognitive-emotional processes underpinning desires to over-consume foods can assist more effective prevention and treatments of obesity. This review addresses recent work linking dietary fat intake and omega-3 polyunsaturated fatty acid dietary imbalance with inflammation in developing, adult, and aged brains. Thus, early-life diet and exposure to stress can lead to cognitive dysfunction throughout life and there is potential for early nutritional interventions (e.g., with essential micronutrients) for preventing these deficits. Likewise, acute consumption of a high-fat diet primes the hippocampus to produce a potentiated neuroinflammatory response to a mild immune challenge, causing memory deficits. Low dietary intake of omega-3 polyunsaturated fatty acids can also contribute to depression through its effects on endocannabinoid and inflammatory pathways in specific brain regions leading to synaptic phagocytosis by microglia in the hippocampus, contributing to memory loss. However, encouragingly, consumption of fruits and vegetables high in polyphenolics can prevent and even reverse age-related cognitive deficits by lowering oxidative stress and inflammation. Understanding relationships between diet, cognition, and emotion is necessary to uncover mechanisms involved in and strategies to prevent or attenuate comorbid neurological conditions in obese individuals.
Collapse
Affiliation(s)
- Sarah J. Spencer
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3788 Australia
| | - Aniko Korosi
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, 1098 XH Netherlands
| | - Sophie Layé
- Nutrition et Neurobiologie Intégrée, INRA, Bordeaux University, Bordeaux, UMR1286 France
| | - Barbara Shukitt-Hale
- USDA-ARS, Human Nutrition Research Center On Aging at Tufts University, Boston, MA 02111-1524 USA
| | - Ruth M. Barrientos
- Department of Psychology & Neuroscience, and Center for Neuroscience, University of Colorado, Campus Box 345, Boulder, CO 80309-0345 USA
| |
Collapse
|
155
|
Heun-Johnson H, Levitt P. Differential impact of Met receptor gene interaction with early-life stress on neuronal morphology and behavior in mice. Neurobiol Stress 2017; 8:10-20. [PMID: 29255778 PMCID: PMC5723381 DOI: 10.1016/j.ynstr.2017.11.003] [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: 08/01/2017] [Revised: 11/24/2017] [Accepted: 11/25/2017] [Indexed: 01/01/2023] Open
Abstract
Early adversity in childhood increases the risk of anxiety, mood, and post-traumatic stress disorders in adulthood, and specific gene-by-environment interactions may increase risk further. A common functional variant in the promoter region of the gene encoding the human MET receptor tyrosine kinase (rs1858830 ‘C’ allele) reduces expression of MET and is associated with altered cortical circuit function and structural connectivity. Mice with reduced Met expression exhibit changes in anxiety-like and conditioned fear behavior, precocious synaptic maturation in the hippocampus, and reduced neuronal arbor complexity and synaptogenesis. These phenotypes also can be produced independently by early adversity in wild-type mice. The present study addresses the outcome of combining early-life stress and genetic influences that alter timing of maturation on enduring functional and structural phenotypes. Using a model of reduced Met expression (Met+/−) and early-life stress from postnatal day 2–9, social, anxiety-like, and contextual fear behaviors in later life were measured. Mice that experienced early-life stress exhibited impairments in social interaction, whereas alterations in anxiety-like behavior and fear learning were driven by Met haploinsufficiency, independent of rearing condition. Early-life stress or reduced Met expression decreased arbor complexity of ventral hippocampal CA1 pyramidal neurons projecting to basolateral amygdala. Paradoxically, arbor complexity in Met+/− mice was increased following early-life stress, and thus not different from arbors in wild-type mice raised in control conditions. The changes in dendritic morphology are consistent with the hypothesis that the physiological state of maturation of CA1 neurons in Met+/− mice influences their responsiveness to early-life stress. The dissociation of behavioral and structural changes suggests that there may be phenotype-specific sensitivities to early-life stress.
Collapse
Affiliation(s)
- Hanke Heun-Johnson
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Pat Levitt
- Institute for the Developing Mind, Children's Hospital Los Angeles, Los Angeles, CA, USA.,Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
156
|
Liang L, Zhou H, Zhang S, Yuan J, Wu H. Effects of gut microbiota disturbance induced in early life on the expression of extrasynaptic GABA-A receptor α5 and δ subunits in the hippocampus of adult rats. Brain Res Bull 2017; 135:113-119. [DOI: 10.1016/j.brainresbull.2017.09.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 09/21/2017] [Accepted: 09/26/2017] [Indexed: 12/26/2022]
|
157
|
Bick J, Zeanah CH, Fox NA, Nelson CA. Memory and Executive Functioning in 12-Year-Old Children With a History of Institutional Rearing. Child Dev 2017; 89:495-508. [PMID: 28898388 DOI: 10.1111/cdev.12952] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
We examined visual recognition memory and executive functioning (spatial working memory [SWM], spatial planning, rule learning, and attention shifting) in 12-year-olds (n = 150) who participated in the Bucharest Early Intervention Project, a randomized controlled trial of foster care for institutionally reared children. Similar to prior reports at 8 years of age, institutionally reared children showed significant deficits in visual recognition memory and SWM. Deficits in attention shifting and rule learning were also apparent at this time point. These data suggest that early experiences continue to shape the development of memory, learning, and executive functioning processes in preadolescence, which may explain broader cognitive and learning difficulties commonly associated with severe early life neglect.
Collapse
Affiliation(s)
- Johanna Bick
- Boston Children's Hospital.,Harvard Medical School
| | | | | | - Charles A Nelson
- Boston Children's Hospital.,Harvard Medical School.,Harvard Graduate School of Education
| |
Collapse
|
158
|
Walker CD, Bath KG, Joels M, Korosi A, Larauche M, Lucassen PJ, Morris MJ, Raineki C, Roth TL, Sullivan RM, Taché Y, Baram TZ. Chronic early life stress induced by limited bedding and nesting (LBN) material in rodents: critical considerations of methodology, outcomes and translational potential. Stress 2017; 20:421-448. [PMID: 28617197 PMCID: PMC5705407 DOI: 10.1080/10253890.2017.1343296] [Citation(s) in RCA: 222] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 06/07/2017] [Accepted: 06/09/2017] [Indexed: 02/07/2023] Open
Abstract
The immediate and long-term effects of exposure to early life stress (ELS) have been documented in humans and animal models. Even relatively brief periods of stress during the first 10 days of life in rodents can impact later behavioral regulation and the vulnerability to develop adult pathologies, in particular an impairment of cognitive functions and neurogenesis, but also modified social, emotional, and conditioned fear responses. The development of preclinical models of ELS exposure allows the examination of mechanisms and testing of therapeutic approaches that are not possible in humans. Here, we describe limited bedding and nesting (LBN) procedures, with models that produce altered maternal behavior ranging from fragmentation of care to maltreatment of infants. The purpose of this paper is to discuss important issues related to the implementation of this chronic ELS procedure and to describe some of the most prominent endpoints and consequences, focusing on areas of convergence between laboratories. Effects on the hypothalamic-pituitary adrenal (HPA) axis, gut axis and metabolism are presented in addition to changes in cognitive and emotional functions. Interestingly, recent data have suggested a strong sex difference in some of the reported consequences of the LBN paradigm, with females being more resilient in general than males. As both the chronic and intermittent variants of the LBN procedure have profound consequences on the offspring with minimal external intervention from the investigator, this model is advantageous ecologically and has a large translational potential. In addition to the direct effect of ELS on neurodevelopmental outcomes, exposure to adverse early environments can also have intergenerational impacts on mental health and function in subsequent generation offspring. Thus, advancing our understanding of the effect of ELS on brain and behavioral development is of critical concern for the health and wellbeing of both the current population, and for generations to come.
Collapse
Affiliation(s)
- Claire-Dominique Walker
- Department of Psychiatry, McGill University, Douglas Mental Health University Institute, 6875 Lasalle Blvd, Montreal, QC H4H 1R3, Canada
| | - Kevin G. Bath
- Department of Neuroscience, Brown University, 185 Meeting Street, Providence, RI 02912, USA
| | - Marian Joels
- Department Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands
| | - Aniko Korosi
- Brain Plasticity group, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098XH, Amsterdam, The Netherlands
| | - Muriel Larauche
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California Los Angeles, and VA Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
| | - Paul J. Lucassen
- Brain Plasticity group, Swammerdam Institute for Life Sciences, University of Amsterdam, Science Park 904, 1098XH, Amsterdam, The Netherlands
| | - Margaret J. Morris
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney 2052, NSW, Australia
| | - Charlis Raineki
- Department of Cellular and Physiological Sciences, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, Department of Child and Adolescent Psychiatry, New York University Langone Medical School, New York, NY 10016, USA
| | - Tania L. Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, USA
| | - Regina M. Sullivan
- Emotional Brain Institute, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, Department of Child and Adolescent Psychiatry, New York University Langone Medical School, New York, NY 10016, USA
| | - Yvette Taché
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California Los Angeles, and VA Greater Los Angeles Healthcare System, Los Angeles, CA, 90073, USA
| | - Tallie Z. Baram
- Department of Pediatrics, of Anatomy & Neurobiology and of Neurology, University of California-Irvine, Irvine, CA 92697, USA
| |
Collapse
|
159
|
Nectin-3 modulates the structural plasticity of dentate granule cells and long-term memory. Transl Psychiatry 2017; 7:e1228. [PMID: 28872640 PMCID: PMC5639241 DOI: 10.1038/tp.2017.196] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 07/05/2017] [Accepted: 07/14/2017] [Indexed: 11/29/2022] Open
Abstract
Nectin-3, a cell adhesion molecule enriched in hippocampal neurons, has been implicated in stress-related cognitive disorders. Nectin-3 is expressed by granule cells in the dentate gyrus (DG), but it remains unclear whether nectin-3 in DG modulates the structural plasticity of dentate granule cells and hippocampus-dependent memory. In this study, we found that DG nectin-3 expression levels were developmentally regulated and reduced by early postnatal stress exposure in adult mice. Most importantly, knockdown of nectin-3 levels in all DG neuron populations by adeno-associated virus (AAV) mimicked the cognitive effects of early-life stress, and impaired long-term spatial memory and temporal order memory. Moreover, AAV-mediated DG nectin-3 knockdown increased the density of doublecortin-immunoreactive differentiating cells under proliferation and calretinin-immunoreactive immature neurons, but markedly decreased calbindin immunoreactivity, indicating that nectin-3 modulates the differentiation and maturation of adult-born DG granule cells. Using retrovirus to target newly generated DG neurons, we found that selective nectin-3 knockdown in new DG neurons also impaired long-term spatial memory. In addition, suppressing nectin-3 expression in new DG neurons evoked a reduction of dendritic spines, especially thin spines. Our data indicate that nectin-3 expressed in DG neurons may modulate adult neurogenesis, dendritic spine plasticity and the cognitive effects of early-life stress.
Collapse
|
160
|
Rosinger ZJ, Jacobskind JS, Park SG, Justice NJ, Zuloaga DG. Distribution of corticotropin-releasing factor receptor 1 in the developing mouse forebrain: A novel sex difference revealed in the rostral periventricular hypothalamus. Neuroscience 2017; 361:167-178. [PMID: 28823817 DOI: 10.1016/j.neuroscience.2017.08.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 07/19/2017] [Accepted: 08/09/2017] [Indexed: 12/16/2022]
Abstract
Corticotropin-releasing factor (CRF) signaling through CRF receptor 1 (CRFR1) regulates autonomic, endocrine and behavioral responses to stress and has been implicated in the pathophysiology of several disorders including anxiety, depression, and addiction. Using a validated CRFR1 reporter mouse line (bacterial artificial chromosome identified by green fluorescence protein (BAC GFP-CRFR1)), we investigated the distribution of CRFR1 in the developing mouse forebrain. Distribution of CRFR1 was investigated at postnatal days (P) 0, 4, and 21 in male and female mice. CRFR1 increased with age in several regions including the medial amygdala, arcuate nucleus, paraventricular hypothalamus, medial septum, CA1 hippocampal area, and the lateral habenula. Regions showing decreased CRFR1 expression with increased age include the intermediate portion of the periventricular hypothalamic nucleus, and CA3 hippocampal area. We report a sexually dimorphic expression of CRFR1 within the rostral portion of the anteroventral periventricular nucleus of the hypothalamus (AVPV/PeN), a region known to regulate ovulation, reproductive and maternal behaviors. Females had a greater number of CRFR1-GFP-ir cells at all time points in the AVPV/PeN and CRFR1-GFP-ir was nearly absent in males by P21. Overall, alterations in CRFR1-GFP-ir distribution based on age and sex may contribute to observed age- and sex-dependent differences in stress regulation.
Collapse
Affiliation(s)
| | | | - Shannon G Park
- University at Albany, Department of Psychology, Albany, NY 12222, USA
| | - Nicholas J Justice
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Sciences Center, Houston, TX, USA
| | - Damian G Zuloaga
- University at Albany, Department of Psychology, Albany, NY 12222, USA.
| |
Collapse
|
161
|
Caregiver maltreatment causes altered neuronal DNA methylation in female rodents. Dev Psychopathol 2017; 29:477-489. [PMID: 28401839 DOI: 10.1017/s0954579417000128] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Negative experiences with a caregiver during infancy can result in long-term changes in brain function and behavior, but underlying mechanisms are not well understood. It is our central hypothesis that brain and behavior changes are conferred by early childhood adversity through epigenetic changes involving DNA methylation. Using a rodent model of early-life caregiver maltreatment (involving exposure to an adverse caregiving environment for postnatal days 1-7), we have previously demonstrated abnormal methylation of DNA associated with the brain-derived neurotrophic factor (Bdnf) gene in the medial prefrontal cortex (mPFC) of adult rats. The aim of the current study was to characterize Bdnf DNA methylation in specific cell populations within the mPFC. In the prefrontal cortex, there is approximately twice as many neurons as glia, and studies have recently shown differential and distinctive DNA methylation patterns in neurons versus nonneurons. Here, we extracted nuclei from the mPFC of adult animals that had experienced maltreatment and used fluorescence-activated cell sorting to isolate cell types before performing bisulfite sequencing to estimate methylation of cytosine-guanine sites. Our data indicate that early-life stress induced methylation of DNA associated with Bdnf IV in a cell-type and sex-specific manner. Specifically, females that experienced early-life maltreatment exhibited greater neuronal cytosine-guanine methylation compared to controls, while no changes were detected in Bdnf methylation in males regardless of cell type. These changes localize the specificity of our previous findings to mPFC neurons and highlight the capacity of maltreatment to cause methylation changes that are likely to have functional consequences for neuronal function.
Collapse
|
162
|
Gutierrez A, Jablonski SA, Amos-Kroohs RM, Barnes AC, Williams MT, Vorhees CV. Effects of Housing on Methamphetamine-Induced Neurotoxicity and Spatial Learning and Memory. ACS Chem Neurosci 2017; 8:1479-1489. [PMID: 28287691 DOI: 10.1021/acschemneuro.6b00419] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Severe stress potentiates methamphetamine (MA) neurotoxicity. However, whether moderate stress increases or decreases the neurotoxic effects of MA is unknown. We assessed the effects of MA (4 × 10 mg/kg at 2 h intervals) in combination with prior barren-cage housing in adult male Sprague-Dawley rats on monoamines and glial fibrillary acid protein (GFAP) in one cohort and spatial learning and memory in the Morris water maze in another cohort. MA reduced dopamine (DA) and serotonin (5-HT) in the neostriatum and nucleus accumbens, 5-HT in the hippocampus, and increased GFAP in neostriatum and nucleus accumbens compared with saline controls. In neostriatum, barren-cage housing protected against MA-induced increases in GFAP, but it did not prevent DA and 5-HT reductions, although it did increase hippocampal norepinephrine. MA impaired spatial learning during acquisition, reversal, and shift phases and impaired reference memory on reversal and shift probe trials. Barren-cage housing enhanced performance during acquisition but not during reversal or shift or on probe trials. The data indicate that prior barren-cage housing moderates MA-induced neostriatal astrogliosis and initial spatial learning, but has no protective effect when the platform is smaller and relocated and therefore requires cognitive flexibility in relearning.
Collapse
Affiliation(s)
- Arnold Gutierrez
- Department of Pediatrics,
Division of Neurology, Cincinnati Children’s Research Foundation, Cincinnati Ohio 45229, United States
- University of Cincinnati College of Medicine, Cincinnati Ohio 45229, United States
| | - Sarah A. Jablonski
- Department of Pediatrics,
Division of Neurology, Cincinnati Children’s Research Foundation, Cincinnati Ohio 45229, United States
| | - Robyn M. Amos-Kroohs
- Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081, United States
| | - Anna C. Barnes
- Department of Pediatrics,
Division of Neurology, Cincinnati Children’s Research Foundation, Cincinnati Ohio 45229, United States
- College
of Arts and Sciences, Cincinnati, University of Cincinnati, Cincinnati Ohio 45229, United States
| | - Michael T. Williams
- Department of Pediatrics,
Division of Neurology, Cincinnati Children’s Research Foundation, Cincinnati Ohio 45229, United States
- University of Cincinnati College of Medicine, Cincinnati Ohio 45229, United States
| | - Charles V. Vorhees
- Department of Pediatrics,
Division of Neurology, Cincinnati Children’s Research Foundation, Cincinnati Ohio 45229, United States
- University of Cincinnati College of Medicine, Cincinnati Ohio 45229, United States
| |
Collapse
|
163
|
Converging, Synergistic Actions of Multiple Stress Hormones Mediate Enduring Memory Impairments after Acute Simultaneous Stresses. J Neurosci 2017; 36:11295-11307. [PMID: 27807170 DOI: 10.1523/jneurosci.2542-16.2016] [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: 08/10/2016] [Accepted: 09/16/2016] [Indexed: 11/21/2022] Open
Abstract
Stress influences memory, an adaptive process crucial for survival. During stress, hippocampal synapses are bathed in a mixture of stress-released molecules, yet it is unknown whether or how these interact to mediate the effects of stress on memory. Here, we demonstrate novel synergistic actions of corticosterone and corticotropin-releasing hormone (CRH) on synaptic physiology and dendritic spine structure that mediate the profound effects of acute concurrent stresses on memory. Spatial memory in mice was impaired enduringly after acute concurrent stresses resulting from loss of synaptic potentiation associated with disrupted structure of synapse-bearing dendritic spines. Combined application of the stress hormones corticosterone and CRH recapitulated the physiological and structural defects provoked by acute stresses. Mechanistically, corticosterone and CRH, via their cognate receptors, acted synergistically on the spine-actin regulator RhoA, promoting its deactivation and degradation, respectively, and destabilizing spines. Accordingly, blocking the receptors of both hormones, but not each alone, rescued memory. Therefore, the synergistic actions of corticosterone and CRH at hippocampal synapses underlie memory impairments after concurrent and perhaps also single, severe acute stresses, with potential implications to spatial memory dysfunction in, for example, posttraumatic stress disorder. SIGNIFICANCE STATEMENT Stress influences memory, an adaptive process crucial for survival. During stress, adrenal corticosterone and hippocampal corticotropin-releasing hormone (CRH) permeate memory-forming hippocampal synapses, yet it is unknown whether (and how) these hormones interact to mediate effects of stress. Here, we demonstrate novel synergistic actions of corticosterone and CRH on hippocampal synaptic plasticity and spine structure that mediate the memory-disrupting effects of stress. Combined application of both hormones provoked synaptic function collapse and spine disruption. Mechanistically, corticosterone and CRH synergized at the spine-actin regulator RhoA, promoting its deactivation and degradation, respectively, and destabilizing spines. Notably, blocking both hormones, but not each alone, prevented the enduring memory problems after acute concurrent stresses. Therefore, synergistic actions of corticosterone and CRH underlie enduring memory impairments after concurrent acute stresses, which might be relevant to spatial memory deficits described in posttraumatic stress disorder.
Collapse
|
164
|
Kanatsou S, Karst H, Kortesidou D, van den Akker RA, den Blaauwen J, Harris AP, Seckl JR, Krugers HJ, Joels M. Overexpression of Mineralocorticoid Receptors in the Mouse Forebrain Partly Alleviates the Effects of Chronic Early Life Stress on Spatial Memory, Neurogenesis and Synaptic Function in the Dentate Gyrus. Front Cell Neurosci 2017; 11:132. [PMID: 28611594 PMCID: PMC5447008 DOI: 10.3389/fncel.2017.00132] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 04/19/2017] [Indexed: 11/13/2022] Open
Abstract
Evidence from human studies suggests that high expression of brain mineralocorticoid receptors (MR) may promote resilience against negative consequences of stress exposure, including childhood trauma. We examined, in mice, whether brain MR overexpression can alleviate the effects of chronic early life stress (ELS) on contextual memory formation under low and high stress conditions, and neurogenesis and synaptic function of dentate gyrus granular cells. Male mice were exposed to ELS by housing the dam with limited nesting and bedding material from postnatal day (PND) 2 to 9. We investigated the moderating role of MRs by using forebrain-specific transgenic MR overexpression (MR-tg) mice. Low-stress contextual (i.e., object relocation) memory formation was hampered by ELS in wildtype but not MR-tg mice. Anxiety like behavior and high-stress contextual (i.e., fear) memory formation were unaffected by ELS and/or MR expression level. At the cellular level, an interaction effect was observed between ELS and MR overexpression on the number of doublecortin-positive cells, with a significant difference between the wildtype ELS and MR-tg ELS groups. No interaction was found regarding Ki-67 and BrdU staining. A significant interaction between ELS and MR expression was further observed with regard to mEPSCs and mIPSC frequency. The ratio of evoked EPSC/IPSC or NMDA/AMPA responses was unaffected. Overall, these results suggest that ELS affects contextual memory formation under low stress conditions as well as neurogenesis and synaptic transmission in dentate granule cells, an effect that can be alleviated by MR-overexpression.
Collapse
Affiliation(s)
- Sofia Kanatsou
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, Netherlands.,Swammerdam Institute for Life Sciences - Center for Neuroscience, University of AmsterdamAmsterdam, Netherlands
| | - Henk Karst
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, Netherlands
| | - Despoina Kortesidou
- Swammerdam Institute for Life Sciences - Center for Neuroscience, University of AmsterdamAmsterdam, Netherlands
| | - Rachelle A van den Akker
- Swammerdam Institute for Life Sciences - Center for Neuroscience, University of AmsterdamAmsterdam, Netherlands
| | - Jan den Blaauwen
- Swammerdam Institute for Life Sciences - Center for Neuroscience, University of AmsterdamAmsterdam, Netherlands
| | - Anjanette P Harris
- Endocrinology Unit, Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of EdinburghEdinburgh, United Kingdom
| | - Jonathan R Seckl
- Endocrinology Unit, Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of EdinburghEdinburgh, United Kingdom
| | - Harm J Krugers
- Swammerdam Institute for Life Sciences - Center for Neuroscience, University of AmsterdamAmsterdam, Netherlands
| | - Marian Joels
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, Netherlands.,University of Groningen, University Medical Center GroningenGroningen, Netherlands
| |
Collapse
|
165
|
Claflin DI, Schmidt KD, Vallandingham ZD, Kraszpulski M, Hennessy MB. Influence of postnatal glucocorticoids on hippocampal-dependent learning varies with elevation patterns and administration methods. Neurobiol Learn Mem 2017; 143:77-87. [PMID: 28545908 DOI: 10.1016/j.nlm.2017.05.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 05/12/2017] [Accepted: 05/16/2017] [Indexed: 11/17/2022]
Abstract
Recent interest in the lasting effects of early-life stress has expanded to include effects on cognitive performance. An increase in circulating glucocorticoids is induced by stress exposure and glucocorticoid effects on the hippocampus likely underlie many of the cognitive consequences. Here we review studies showing that corticosterone administered to young rats at the conclusion of the stress-hyporesponsiveness period affects later performance in hippocampally-mediated trace eyeblink conditioning. The nature and even direction of these effects varies with the elevation patterns (level, duration, temporal fluctuation) achieved by different administration methods. We present new time course data indicating that constant glucocorticoid elevations generally corresponded with hippocampus-mediated learning deficits, whereas acute, cyclical elevations corresponded with improved initial acquisition. Sensitivity was greater for males than for females. Further, changes in hippocampal neurogenesis paralleled some but not all effects. The findings demonstrate that specific patterns of glucocorticoid elevation produced by different drug administration procedures can have markedly different, sex-specific consequences on basic cognitive performance and underlying hippocampal physiology. Implications of these findings for glucocorticoid medications prescribed in childhood are discussed.
Collapse
Affiliation(s)
- Dragana I Claflin
- Department of Psychology, Wright State University, 3640 Colonel Glenn Hwy, Dayton, OH 45435, USA.
| | - Kevin D Schmidt
- Department of Psychology, Wright State University, 3640 Colonel Glenn Hwy, Dayton, OH 45435, USA; Applied Neuroscience Branch, Air Force Research Laboratory, 2510 Fifth St., Wright-Patterson AFB, OH 45433, USA.
| | - Zachary D Vallandingham
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, 3640 Colonel Glenn Hwy, Dayton, OH 45435, USA.
| | - Michal Kraszpulski
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, 3640 Colonel Glenn Hwy, Dayton, OH 45435, USA.
| | - Michael B Hennessy
- Department of Psychology, Wright State University, 3640 Colonel Glenn Hwy, Dayton, OH 45435, USA.
| |
Collapse
|
166
|
van Bodegom M, Homberg JR, Henckens MJAG. Modulation of the Hypothalamic-Pituitary-Adrenal Axis by Early Life Stress Exposure. Front Cell Neurosci 2017; 11:87. [PMID: 28469557 PMCID: PMC5395581 DOI: 10.3389/fncel.2017.00087] [Citation(s) in RCA: 325] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/13/2017] [Indexed: 12/20/2022] Open
Abstract
Exposure to stress during critical periods in development can have severe long-term consequences, increasing overall risk on psychopathology. One of the key stress response systems mediating these long-term effects of stress is the hypothalamic-pituitary-adrenal (HPA) axis; a cascade of central and peripheral events resulting in the release of corticosteroids from the adrenal glands. Activation of the HPA-axis affects brain functioning to ensure a proper behavioral response to the stressor, but stress-induced (mal)adaptation of the HPA-axis' functional maturation may provide a mechanistic basis for the altered stress susceptibility later in life. Development of the HPA-axis and the brain regions involved in its regulation starts prenatally and continues after birth, and is protected by several mechanisms preventing corticosteroid over-exposure to the maturing brain. Nevertheless, early life stress (ELS) exposure has been reported to have numerous consequences on HPA-axis function in adulthood, affecting both its basal and stress-induced activity. According to the match/mismatch theory, encountering ELS prepares an organism for similar ("matching") adversities during adulthood, while a mismatching environment results in an increased susceptibility to psychopathology, indicating that ELS can exert either beneficial or disadvantageous effects depending on the environmental context. Here, we review studies investigating the mechanistic underpinnings of the ELS-induced alterations in the structural and functional development of the HPA-axis and its key external regulators (amygdala, hippocampus, and prefrontal cortex). The effects of ELS appear highly dependent on the developmental time window affected, the sex of the offspring, and the developmental stage at which effects are assessed. Albeit by distinct mechanisms, ELS induced by prenatal stressors, maternal separation, or the limited nesting model inducing fragmented maternal care, typically results in HPA-axis hyper-reactivity in adulthood, as also found in major depression. This hyper-activity is related to increased corticotrophin-releasing hormone signaling and impaired glucocorticoid receptor-mediated negative feedback. In contrast, initial evidence for HPA-axis hypo-reactivity is observed for early social deprivation, potentially reflecting the abnormal HPA-axis function as observed in post-traumatic stress disorder, and future studies should investigate its neural/neuroendocrine foundation in further detail. Interestingly, experiencing additional (chronic) stress in adulthood seems to normalize these alterations in HPA-axis function, supporting the match/mismatch theory.
Collapse
Affiliation(s)
| | | | - Marloes J. A. G. Henckens
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition and BehaviourRadboudumc, Nijmegen, Netherlands
| |
Collapse
|
167
|
Bolton JL, Molet J, Ivy A, Baram TZ. New insights into early-life stress and behavioral outcomes. Curr Opin Behav Sci 2017; 14:133-139. [PMID: 28413813 DOI: 10.1016/j.cobeha.2016.12.012] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Adverse early-life experiences, including various forms of early-life stress, have consistently been linked with vulnerability to cognitive and emotional disorders later in life. Understanding the mechanisms underlying the enduring consequences of early-life stress is an active area of research, because this knowledge is critical for developing potential interventions. Animal models of early-life stress typically rely on manipulating maternal/parental presence and care, because these are the major source of early-life experiences in humans. Diverse models have been created, and have resulted in a wealth of behavioral outcomes. Here we focus on recent findings highlighting early-life stress-induced behavioral disturbances, ranging from hippocampus-dependent memory deficits to problems with experiencing pleasure (anhedonia). The use of naturalistic animal models of chronic early-life stress provides insight into the spectrum of cognitive and emotional outcomes and enables probing the underlying mechanisms using molecular-, cellular-, and network-level approaches.
Collapse
Affiliation(s)
- Jessica L Bolton
- Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, California 92697-4475.,Department of Pediatrics, University of California-Irvine, Irvine, California 92697-4475
| | - Jenny Molet
- Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, California 92697-4475.,Department of Pediatrics, University of California-Irvine, Irvine, California 92697-4475
| | - Autumn Ivy
- Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, California 92697-4475.,Department of Pediatrics, University of California-Irvine, Irvine, California 92697-4475
| | - Tallie Z Baram
- Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, California 92697-4475.,Department of Pediatrics, University of California-Irvine, Irvine, California 92697-4475
| |
Collapse
|
168
|
Effects of early life stress on rodent hippocampal synaptic plasticity: a systematic review. Curr Opin Behav Sci 2017. [DOI: 10.1016/j.cobeha.2017.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
169
|
Effects of Traumatic Stress Induced in the Juvenile Period on the Expression of Gamma-Aminobutyric Acid Receptor Type A Subunits in Adult Rat Brain. Neural Plast 2017; 2017:5715816. [PMID: 28352479 PMCID: PMC5352903 DOI: 10.1155/2017/5715816] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 01/18/2017] [Accepted: 02/15/2017] [Indexed: 12/04/2022] Open
Abstract
Studies have found that early traumatic experience significantly increases the risk of posttraumatic stress disorder (PTSD). Gamma-aminobutyric acid (GABA) deficits were proposed to be implicated in development of PTSD, but the alterations of GABA receptor A (GABAAR) subunits induced by early traumatic stress have not been fully elucidated. Furthermore, previous studies suggested that exercise could be more effective than medications in reducing severity of anxiety and depression but the mechanism is unclear. This study used inescapable foot-shock to induce PTSD in juvenile rats and examined their emotional changes using open-field test and elevated plus maze, memory changes using Morris water maze, and the expression of GABAAR subunits (γ2, α2, and α5) in subregions of the brain in the adulthood using western blotting and immunohistochemistry. We aimed to observe the role of GABAAR subunits changes induced by juvenile trauma in the pathogenesis of subsequent PTSD in adulthood. In addition, we investigated the protective effects of exercise for 6 weeks and benzodiazepine (clonazepam) for 2 weeks. This study found that juvenile traumatic stress induced chronic anxiety and spatial memory loss and reduced expression of GABAAR subunits in the adult rat brains. Furthermore, exercise led to significant improvement as compared to short-term BZ treatment.
Collapse
|
170
|
Adrenocorticotropic hormone protects learning and memory function in epileptic Kcna1-null mice. Neurosci Lett 2017; 645:14-18. [PMID: 28249786 DOI: 10.1016/j.neulet.2017.02.069] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 02/10/2017] [Accepted: 02/25/2017] [Indexed: 11/20/2022]
Abstract
ACTH, a member of the melanocortin family of peptides, is often used in the treatment of the developmental epileptic encephalopathy spectrum disorders including, Ohtahara, West, Lennox Gastaut and Landau-Kleffner Syndromes and electrical status epilepticus of sleep. In these disorders, although ACTH is often successful in controlling the seizures and/or inter-ictal EEG abnormalities, it is unknown whether ACTH possesses other beneficial effects independent of seizure control. We tested whether ACTH can ameliorate the intrinsic impairment of hippocampal-based learning and memory in epileptic Kcna1-null (KO) mice. We found that ACTH - administered in the form of Acthar Gel given i.p. four times daily at a dose of 4 IU/kg (16 IU/kg/day) for 7days - prevented impairment of long-term potentiation (LTP) evoked with high-frequency stimulation in CA1 hippocampus and also restored spatial learning and memory on the Barnes maze test. However, with this treatment regimen, ACTH did not exert a significant effect on the frequency of spontaneous recurrent seizures. Together, our findings indicate that ACTH can ameliorate memory impairment in epileptic Kcna1-null mice separate from seizure control, and suggest that this widely used peptide may exert direct nootropic effects in the epileptic brain.
Collapse
|
171
|
McIlwrick S, Pohl T, Chen A, Touma C. Late-Onset Cognitive Impairments after Early-Life Stress Are Shaped by Inherited Differences in Stress Reactivity. Front Cell Neurosci 2017; 11:9. [PMID: 28261058 PMCID: PMC5306385 DOI: 10.3389/fncel.2017.00009] [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: 09/27/2016] [Accepted: 01/12/2017] [Indexed: 01/18/2023] Open
Abstract
Early-life stress (ELS) has been associated with lasting cognitive impairments and with an increased risk for affective disorders. A dysregulation of the hypothalamus-pituitary-adrenal (HPA) axis, the body’s main stress response system, is critically involved in mediating these long-term consequences of adverse early-life experience. It remains unclear to what extent an inherited predisposition for HPA axis sensitivity or resilience influences the relationship between ELS and cognitive impairments, and which neuroendocrine and molecular mechanisms may be involved. To investigate this, we exposed animals of the stress reactivity mouse model, consisting of three independent lines selectively bred for high (HR), intermediate (IR), or low (LR) HPA axis reactivity to a stressor, to ELS and assessed their cognitive performance, neuroendocrine function and hippocampal gene expression in early and in late adulthood. Our results show that HR animals that were exposed to ELS exhibited an HPA axis hyper-reactivity in early and late adulthood, associated with cognitive impairments in hippocampus-dependent tasks, as well as molecular changes in transcript levels involved in the regulation of HPA axis activity (Crh) and in neurotrophic action (Bdnf). In contrast, LR animals showed intact cognitive function across adulthood, with no change in stress reactivity. Intriguingly, LR animals that were exposed to ELS even showed significant signs of enhanced cognitive performance in late adulthood, which may be related to late-onset changes observed in the expression of Crh and Crhr1 in the dorsal hippocampus of these animals. Collectively, our findings demonstrate that the lasting consequences of ELS at the level of cognition differ as a function of inherited predispositions and suggest that an innate tendency for low stress reactivity may be protective against late-onset cognitive impairments after ELS.
Collapse
Affiliation(s)
- Silja McIlwrick
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry Munich, Germany
| | - Tobias Pohl
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry Munich, Germany
| | - Alon Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of PsychiatryMunich, Germany; Department of Neurobiology, Weizmann Institute of ScienceRehovot, Israel
| | - Chadi Touma
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of PsychiatryMunich, Germany; Department of Behavioural Biology, University of OsnabrückOsnabrück, Germany
| |
Collapse
|
172
|
Early-life stress impairs recognition memory and perturbs the functional maturation of prefrontal-hippocampal-perirhinal networks. Sci Rep 2017; 7:42042. [PMID: 28169319 PMCID: PMC5294456 DOI: 10.1038/srep42042] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 01/06/2017] [Indexed: 12/15/2022] Open
Abstract
Early life exposure to stressful situations impairs cognitive performance of adults and contributes to the etiology of several psychiatric disorders. Most of affected cognitive abilities rely on coupling by synchrony within complex neuronal networks, including prefrontal cortex (PFC), hippocampus (HP), and perirhinal cortex (PRH). Yet it remains poorly understood how early life stress (ELS) induces dysfunction within these networks during the course of development. Here we used intermittent maternal separation during the first 2 postnatal weeks to mimic ELS and monitored the recognition memory and functional coupling within prefrontal-hippocampal-perirhinal circuits in juvenile rats. While maternally-separated female rats showed largely normal behavior, male rats experiencing this form of ELS had poorer location and recency recognition memory. Simultaneous multi-site extracellular recordings of network oscillations and neuronal spiking from PFC, HP, and PRH in vivo revealed corresponding decrease of oscillatory activity in theta and beta frequency bands in the PFC of male but not female rats experiencing maternal separation. This deficit was accompanied by weaker cross-frequency coupling within juvenile prefrontal-hippocampal networks. These results indicate that already at juvenile age ELS mimicked by maternal separation induces sex-specific deficits in recognition memory that might have as underlying mechanism a disturbed communication between PFC and HP.
Collapse
|
173
|
Hippocampal Contribution to Context Encoding across Development Is Disrupted following Early-Life Adversity. J Neurosci 2017; 37:1925-1934. [PMID: 28093475 DOI: 10.1523/jneurosci.2618-16.2017] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 01/07/2017] [Accepted: 01/12/2017] [Indexed: 12/15/2022] Open
Abstract
Context can drastically influence responses to environmental stimuli. For example, a gunshot should provoke a different response at a public park than a shooting range. Little is known about how contextual processing and neural correlates change across human development or about individual differences related to early environmental experiences. Children (N = 60; 8-19 years, 24 exposed to interpersonal violence) completed a context encoding task during fMRI scanning using a delayed match-to-sample design with neutral, happy, and angry facial cues embedded in realistic background scenes. Outside the scanner, participants completed a memory test for context-face pairings. Context memory and neural correlates of context encoding did not vary with age. Larger hippocampal volume was associated with better context memory. Posterior hippocampus was recruited during context encoding, and greater activation in this region predicted better memory for contexts paired with angry faces. Children exposed to violence had poor memory of contexts paired with angry faces, reduced hippocampal volume, and atypical neural recruitment on encoding trials with angry faces, including reduced hippocampal activation and greater functional connectivity between hippocampus and ventrolateral prefrontal cortex (vlPFC). Greater hippocampus-vlPFC connectivity was associated with worse memory for contexts paired with angry faces. Posterior hippocampus appears to support context encoding, a process that does not exhibit age-related variation from middle childhood to late adolescence. Exposure to dangerous environments in childhood is associated with poor context encoding in the presence of threat, likely due to greater vlPFC-dependent attentional narrowing on threat cues at the expense of hippocampus-dependent processing of the broader context.SIGNIFICANCE STATEMENT The ability to use context to guide reactions to environmental stimuli promotes flexible behavior. Remarkably little research has examined how contextual processing changes across development or about influences of the early environment. We provide evidence for posterior hippocampus involvement in context encoding in youth and lack of age-related variation from middle childhood to late adolescence. Children exposed to interpersonal violence exhibited poor memory of contexts paired with angry faces and atypical neural recruitment during context encoding in the presence of threatening facial cues. Heightened attention to threat following violence exposure may come at the expense of encoding contextual information, which may ultimately contribute to pathological fear expressed in safe contexts.
Collapse
|
174
|
Ogura A, Saito S, Kimura S, Tominaga-Yoshino K. [An in vitro model system for studying the stress-induced memory disorder]. Nihon Yakurigaku Zasshi 2017; 150:223-227. [PMID: 29118284 DOI: 10.1254/fpj.150.223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
|
175
|
Wiet S. Origins of Addiction Predictably Embedded in Childhood Trauma: A Neurobiological Review. Soa Chongsonyon Chongsin Uihak 2017. [DOI: 10.5765/jkacap.2017.28.1.4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Affiliation(s)
- Susie Wiet
- General, Child and Adolescent Psychiatrist, Addiction Medicine, Holistic Treament for Psychiatry Trauma Addiction, University of Utah-Adjunct Volunteer Faculty, Salt Lake City, UT, USA
| |
Collapse
|
176
|
Bale TL, Epperson CN. Sex as a Biological Variable: Who, What, When, Why, and How. Neuropsychopharmacology 2017; 42:386-396. [PMID: 27658485 PMCID: PMC5399243 DOI: 10.1038/npp.2016.215] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/22/2016] [Accepted: 09/16/2016] [Indexed: 12/22/2022]
Abstract
The inclusion of sex as a biological variable in research is absolutely essential for improving our understanding of disease mechanisms contributing to risk and resilience. Studies focusing on examining sex differences have demonstrated across many levels of analyses and stages of brain development and maturation that males and females can differ significantly. This review will discuss examples of animal models and clinical studies to provide guidance and reference for the inclusion of sex as an important biological variable relevant to a Neuropsychopharmacology audience.
Collapse
Affiliation(s)
- Tracy L Bale
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn PROMOTES Research on Sex and Gender in Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry,Perelman School of Medicine at the University of Pennsylvania Philadelphia, PA, USA
| | - C Neill Epperson
- Penn PROMOTES Research on Sex and Gender in Health, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychiatry,Perelman School of Medicine at the University of Pennsylvania Philadelphia, PA, USA
- Department of Obstetrics and Gynecology, Perelman School of Medicine at the University of Pennsylvania Philadelphia, PA, USA
| |
Collapse
|
177
|
Wearick-Silva LE, Marshall P, Viola TW, Centeno-Silva A, de Azeredo LA, Orso R, Li X, Donadio MV, Bredy TW, Grassi-Oliveira R. Running during adolescence rescues a maternal separation-induced memory impairment in female mice: Potential role of differential exon-specific BDNF expression. Dev Psychobiol 2016; 59:268-274. [PMID: 27807856 DOI: 10.1002/dev.21487] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 10/19/2016] [Indexed: 12/19/2022]
Abstract
Exposure to early life stress has been associated with memory impairments related to changes in brain-derived neurotrophic factor (BDNF) signaling. However, the potential impact of physical exercise to reverse these effects of maternal separation has been under investigated. Mice were subjected to maternal separation during the first 2 weeks of life and then exposed to a 3-week running protocol during adolescence. The spontaneous object recognition task was performed during adolescence followed by analysis of hippocampal expression of exons I, IV, and IX of the BDNF gene. As expected, maternal separation impaired recognition memory and this effect was reversed by exercise. In addition, running increased BDNF exon I expression, but decreased expression of BDNF exon IV in all groups, while exon IX expression increased only in MS animals exposed to exercise. Our data suggest that memory deficits can be attenuated by exercise and specific transcripts of the BDNF gene are dynamically regulated following both MS and exercise.
Collapse
Affiliation(s)
- Luis Eduardo Wearick-Silva
- Graduate Program in Pediatrics and Child Health, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil.,Developmental Cognitive Neuroscience Laboratory (DCNL) Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Paul Marshall
- Department of Neurobiology and Behavior, University of California - Irvine, Irvine, California
| | - Thiago Wendt Viola
- Graduate Program in Pediatrics and Child Health, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil.,Developmental Cognitive Neuroscience Laboratory (DCNL) Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Anderson Centeno-Silva
- Graduate Program in Pediatrics and Child Health, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil.,Developmental Cognitive Neuroscience Laboratory (DCNL) Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Lucas Araújo de Azeredo
- Graduate Program in Pediatrics and Child Health, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil.,Developmental Cognitive Neuroscience Laboratory (DCNL) Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Rodrigo Orso
- Graduate Program in Pediatrics and Child Health, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil.,Developmental Cognitive Neuroscience Laboratory (DCNL) Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Xiang Li
- Department of Neurobiology and Behavior, University of California - Irvine, Irvine, California
| | - Márcio V Donadio
- Graduate Program in Pediatrics and Child Health, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Timothy W Bredy
- Department of Neurobiology and Behavior, University of California - Irvine, Irvine, California
| | - Rodrigo Grassi-Oliveira
- Graduate Program in Pediatrics and Child Health, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Rio Grande do Sul, Brazil.,Developmental Cognitive Neuroscience Laboratory (DCNL) Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| |
Collapse
|
178
|
Molet J, Maras PM, Kinney-Lang E, Harris NG, Rashid F, Ivy AS, Solodkin A, Obenaus A, Baram TZ. MRI uncovers disrupted hippocampal microstructure that underlies memory impairments after early-life adversity. Hippocampus 2016; 26:1618-1632. [PMID: 27657911 DOI: 10.1002/hipo.22661] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2016] [Indexed: 12/13/2022]
Abstract
Memory and related cognitive functions are progressively impaired in a subgroup of individuals experiencing childhood adversity and stress. However, it is not possible to identify vulnerable individuals early, a crucial step for intervention. In this study, high-resolution magnetic resonance imaging (MRI) and intra-hippocampal diffusion tensor imaging (DTI) were employed to examine for structural signatures of cognitive adolescent vulnerabilities in a rodent model of early-life adversity. These methods were complemented by neuroanatomical and functional assessments of hippocampal network integrity during adolescence, adulthood and middle-age. The high-resolution MRI identified selective loss of dorsal hippocampal volume, and intra-hippocampal DTI uncovered disruption of dendritic structure, consistent with disrupted local connectivity, already during late adolescence in adversity-experiencing rats. Memory deteriorated over time, and stunting of hippocampal dendritic trees was apparent on neuroanatomical analyses. Thus, disrupted hippocampal neuronal structure and connectivity, associated with cognitive impairments, are detectable via non-invasive imaging modalities in rats experiencing early-life adversity. These high-resolution imaging approaches may constitute promising tools for prediction and assessment of at-risk individuals in the clinic. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Pamela M Maras
- Department of Pediatrics, UC-Irvine, Irvine, CA, 92697-4475, USA
| | - Eli Kinney-Lang
- Department of Pediatrics, UC-Irvine, Irvine, CA, 92697-4475, USA.,Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Neil G Harris
- Department of Neurosurgery, UCLA, Los Angeles, CA, 90095-6901, USA
| | - Faisal Rashid
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | | | - Ana Solodkin
- Department of Anatomy and Neurobiology.,Department of Neurology, UC-Irvine, Irvine, CA, 92697-4475, USA
| | - Andre Obenaus
- Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Tallie Z Baram
- Department of Anatomy and Neurobiology.,Department of Pediatrics, UC-Irvine, Irvine, CA, 92697-4475, USA.,Department of Neurology, UC-Irvine, Irvine, CA, 92697-4475, USA
| |
Collapse
|
179
|
Delpech JC, Wei L, Hao J, Yu X, Madore C, Butovsky O, Kaffman A. Early life stress perturbs the maturation of microglia in the developing hippocampus. Brain Behav Immun 2016; 57:79-93. [PMID: 27301858 PMCID: PMC5010940 DOI: 10.1016/j.bbi.2016.06.006] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 05/24/2016] [Accepted: 06/11/2016] [Indexed: 12/22/2022] Open
Abstract
Children exposed to abuse or neglect show abnormal hippocampal development and similar findings have been reported in rodent models. Using brief daily separation (BDS), a mouse model of early life stress, we previously showed that exposure to BDS impairs hippocampal function in adulthood and perturbs synaptic maturation, synaptic pruning, axonal growth and myelination in the developing hippocampus. Given that microglia are involved in these developmental processes, we tested whether BDS impairs microglial activity in the hippocampus of 14 (during BDS) and 28-day old mice (one week after BDS). We found that BDS increased the density and altered the morphology of microglia in the hippocampus of 14-day old pups, effects that were no longer present on postnatal day (PND) 28. Despite the normal cell number and morphology seen at PND28, the molecular signature of hippocampal microglia, assessed using the NanoString immune panel, was altered at both ages. We showed that during normal hippocampal development, microglia undergo significant changes between PND14 and PND28, including reduced cell density, decreased ex vivo phagocytic activity, and an increase in the expression of genes involved in inflammation and cell migration. However, microglia harvested from the hippocampus of 28-day old BDS mice showed an increase in phagocytic activity and reduced expression of genes that normally increase across development. Promoter analysis indicated that alteration in the transcriptional activity of PU.1, Creb1, Sp1, and RelA accounted for most of the transcriptional changes seen during normal microglia development and for most of the BDS-induced changes at PND14 and PND28. These findings are the first to demonstrate that early life stress dysregulates microglial function in the developing hippocampus and to identify key transcription factors that are likely to mediate these changes.
Collapse
Affiliation(s)
- Jean-Christophe Delpech
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA
| | - Lan Wei
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA
| | - Jin Hao
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA
| | - Xiaoqing Yu
- W.M. Keck Foundation Biotechnology Resource Laboratory, Yale University, New Haven, CT 06511, USA
| | - Charlotte Madore
- Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Oleg Butovsky
- Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA.
| |
Collapse
|
180
|
Chaby LE. Why are there lasting effects from exposure to stress during development? An analysis of current models of early stress. Physiol Behav 2016; 164:164-81. [DOI: 10.1016/j.physbeh.2016.05.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/29/2016] [Accepted: 05/17/2016] [Indexed: 01/19/2023]
|
181
|
Insight from animal models of environmentally driven epigenetic changes in the developing and adult brain. Dev Psychopathol 2016; 28:1229-1243. [PMID: 27687803 DOI: 10.1017/s095457941600081x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The efforts of many neuroscientists are directed toward understanding the appreciable plasticity of the brain and behavior. In recent years, epigenetics has become a core of this focus as a prime mechanistic candidate for behavioral modifications. Animal models have been instrumental in advancing our understanding of environmentally driven changes to the epigenome in the developing and adult brain. This review focuses mainly on such discoveries driven by adverse environments along with their associated behavioral outcomes. While much of the evidence discussed focuses on epigenetics within the central nervous system, several peripheral studies in humans who have experienced significant adversity are also highlighted. As we continue to unravel the link between epigenetics and phenotype, discerning the complexity and specificity of epigenetic changes induced by environments is an important step toward understanding optimal development and how to prevent or ameliorate behavioral deficits bred by disruptive environments.
Collapse
|
182
|
Henckens MJAG, Deussing JM, Chen A. Region-specific roles of the corticotropin-releasing factor-urocortin system in stress. Nat Rev Neurosci 2016; 17:636-51. [PMID: 27586075 DOI: 10.1038/nrn.2016.94] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Dysregulation of the corticotropin-releasing factor (CRF)-urocortin (UCN) system has been implicated in stress-related psychopathologies such as depression and anxiety. It has been proposed that CRF-CRF receptor type 1 (CRFR1) signalling promotes the stress response and anxiety-like behaviour, whereas UCNs and CRFR2 activation mediate stress recovery and the restoration of homeostasis. Recent findings, however, provide clear evidence that this view is overly simplistic. Instead, a more complex picture has emerged that suggests that there are brain region- and cell type-specific effects of CRFR signalling that are influenced by the individual's prior experience and that shape molecular, cellular and ultimately behavioural responses to stressful challenges.
Collapse
Affiliation(s)
- Marloes J A G Henckens
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel.,Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany.,Department of Cognitive Neuroscience, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Centre, 6500 HB Nijmegen, The Netherlands
| | - Jan M Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Alon Chen
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 7610001, Israel.,Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| |
Collapse
|
183
|
Amos-Kroohs RM, Davenport LL, Gutierrez A, Hufgard JR, Vorhees CV, Williams MT. Developmental manganese exposure in combination with developmental stress and iron deficiency: Effects on behavior and monoamines. Neurotoxicol Teratol 2016; 56:55-67. [PMID: 27302314 DOI: 10.1016/j.ntt.2016.06.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/09/2016] [Accepted: 06/10/2016] [Indexed: 12/20/2022]
Abstract
Manganese (Mn) is an essential element but neurotoxic at higher exposures, however, Mn exposure seldom occurs in isolation. It often co-occurs in populations with inadequate dietary iron (Fe) and limited resources that result in stress. Subclinical FeD affects up to 15% of U.S. children and exacerbates Mn toxicity by increasing Mn bioavailability. Therefore, we investigated Mn overexposure (MnOE) in rats in combination with Fe deficiency (FeD) and developmental stress, for which we used barren cage rearing. For barren cage rearing (BAR), rats were housed in cages with a wire grid floor or standard bedding material (STD) from embryonic day (E)7 through postnatal day (P)28. For FeD, dams were fed a 90% Fe-deficient NIH-07 diet from E15 through P28. Within each litter, different offspring were treated with 100mg/kg Mn (MnOE) or vehicle (VEH) by gavage every other day from P4-28. Behavior was assessed at two ages and consisted of: open-field, anxiety tests, acoustic startle response (ASR) with prepulse inhibition (PPI), sociability, sucrose preference, tapered beam crossing, and the Porsolt's forced swim test. MnOE had main effects of decreasing activity, ASR, social preference, and social novelty. BAR and FeD transiently modified MnOE effects. BAR groups weighed less and showed decreased anxiety in the elevated zero maze, had increased ASR and decreased PPI, and exhibited reduced sucrose preference compared with the STD groups. FeD animals also weighed less and had increased slips on the tapered beam. Most of the monoamine effects were dopaminergic and occurred in the MnOE groups. The results showed that Mn is a pervasive developmental neurotoxin, the effects of which are modulated by FeD and/or BAR cage rearing.
Collapse
Affiliation(s)
- Robyn M Amos-Kroohs
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, United States; University of Cincinnati College of Medicine, Cincinnati, OH 45229, United States
| | - Laurie L Davenport
- Department of Environmental Health, University of Cincinnati, 3223 Eden Ave., Cincinnati, OH 45220, United States
| | - Arnold Gutierrez
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, United States; University of Cincinnati College of Medicine, Cincinnati, OH 45229, United States
| | - Jillian R Hufgard
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, United States; University of Cincinnati College of Medicine, Cincinnati, OH 45229, United States
| | - Charles V Vorhees
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, United States; University of Cincinnati College of Medicine, Cincinnati, OH 45229, United States
| | - Michael T Williams
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, United States; University of Cincinnati College of Medicine, Cincinnati, OH 45229, United States.
| |
Collapse
|
184
|
Morris MJ, Le V, Maniam J. The impact of poor diet and early life stress on memory status. Curr Opin Behav Sci 2016. [DOI: 10.1016/j.cobeha.2016.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
185
|
Bath K, Manzano-Nieves G, Goodwill H. Early life stress accelerates behavioral and neural maturation of the hippocampus in male mice. Horm Behav 2016; 82:64-71. [PMID: 27155103 PMCID: PMC5308418 DOI: 10.1016/j.yhbeh.2016.04.010] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 04/20/2016] [Accepted: 04/23/2016] [Indexed: 12/22/2022]
Abstract
Early life stress (ELS) increases the risk for later cognitive and emotional dysfunction. ELS is known to truncate neural development through effects on suppressing cell birth, increasing cell death, and altering neuronal morphology, effects that have been associated with behavioral profiles indicative of precocious maturation. However, how earlier silencing of growth drives accelerated behavioral maturation has remained puzzling. Here, we test the novel hypothesis that, ELS drives a switch from growth to maturation to accelerate neural and behavioral development. To test this, we used a mouse model of ELS, fragmented maternal care, and a cross-sectional dense sampling approach focusing on hippocampus and measured effects of ELS on the ontogeny of behavioral development and biomarkers of neural maturation. Consistent with previous work, ELS was associated with an earlier developmental decline in expression of markers of cell proliferation (Ki-67) and differentiation (doublecortin). However, ELS also led to a precocious arrival of Parvalbumin-positive cells, led to an earlier switch in NMDA receptor subunit expression (marker of synaptic maturity), and was associated with an earlier rise in myelin basic protein expression (key component of the myelin sheath). In addition, in a contextual fear-conditioning task, ELS accelerated the timed developmental suppression of contextual fear. Together, these data provide support for the hypothesis that ELS serves to switch neurodevelopment from processes of growth to maturation and promotes accelerated development of some forms of emotional learning.
Collapse
Affiliation(s)
- K Bath
- Department of Cognitive, Linguistic, and Psychological Sciences, Brown University, Providence, RI 02912, United States.
| | - G Manzano-Nieves
- Department of Neuroscience, Brown University, Providence, RI, 02912, United States
| | - H Goodwill
- Department of Neuroscience, Brown University, Providence, RI, 02912, United States
| |
Collapse
|
186
|
Holschneider DP, Guo Y, Mayer EA, Wang Z. Early life stress elicits visceral hyperalgesia and functional reorganization of pain circuits in adult rats. Neurobiol Stress 2016; 3:8-22. [PMID: 26751119 PMCID: PMC4700548 DOI: 10.1016/j.ynstr.2015.12.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Early life stress (ELS) is a risk factor for developing functional gastrointestinal disorders, and has been proposed to be related to a central amplification of sensory input and resultant visceral hyperalgesia. We sought to characterize ELS-related changes in functional brain responses during acute noxious visceral stimulation. Neonatal rats (males/females) were exposed to limited bedding (ELS) or standard bedding (controls) on postnatal days 2–9. Age 10–11 weeks, animals were implanted with venous cannulas and transmitters for abdominal electromyography (EMG). Cerebral blood flow (rCBF) was mapped during colorectal distension (CRD) using [14C]-iodoantipyrine autoradiography, and analyzed in three-dimensionally reconstructed brains by statistical parametric mapping and functional connectivity. EMG responses to CRD were increased after ELS, with no evidence of a sex difference. ELS rats compared to controls showed a greater significant positive correlation of EMG with amygdalar rCBF. Factorial analysis revealed a significant main effect of ‘ELS’ on functional activation of nodes within the pain pathway (somatosensory, insular, cingulate and prefrontal cortices, locus coeruleus/lateral parabrachial n. [LC/LPB], periaqueductal gray, sensory thalamus), as well as in the amygdala, hippocampus and hypothalamus. In addition, ELS resulted in an increase in the number of significant functional connections (i.e. degree centrality) between regions within the pain circuit, including the amygdala, LC/LPB, insula, anterior ventral cingulate, posterior cingulate (retrosplenium), and stria terminalis, with decreases noted in the sensory thalamus and the hippocampus. Sex differences in rCBF were less broadly expressed, with significant differences noted at the level of the cortex, amygdala, dorsal hippocampus, raphe, sensory thalamus, and caudate-putamen. ELS showed a sexually dimorphic effect (‘Sex x ELS’ interaction) at the LC/LPB complex, globus pallidus, hypothalamus, raphe, septum, caudate-putamen and cerebellum. Our results suggest that ELS alters functional activation of the thalamo-cortico-amydala pathway, as well as the emotional-arousal network (amygdala, locus coeruleus), with evidence that ELS may additionally show sexually dimorphic effects on brain function. Early life stress (ELS) elicits visceral hyperalgesia in adult offspring. ELS alters functional activation of the thalamo-cortico-amydala pathway. ELS shows a sexually dimorphic effects on brain function. Functional imaging-based endpoints promise improved animal-to-human translation.
Collapse
Affiliation(s)
- D P Holschneider
- Department of Psychiatry and Behavioral Sciences, University of Southern California, Los Angeles, CA, USA; Departments of Neurology, Cell and Neurobiology, Biomedical Engineering, University of Southern California, Los Angeles, CA, USA; Center for Neurobiology of Stress, Department of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Y Guo
- Department of Psychiatry and Behavioral Sciences, University of Southern California, Los Angeles, CA, USA
| | - E A Mayer
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California, United States of America; Center for Neurobiology of Stress, Department of Medicine, University of California Los Angeles, Los Angeles, California, USA; Departments of Physiology, Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, California, USA
| | - Z Wang
- Department of Psychiatry and Behavioral Sciences, University of Southern California, Los Angeles, CA, USA; Center for Neurobiology of Stress, Department of Medicine, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
187
|
Maniam J, Antoniadis CP, Le V, Morris MJ. A diet high in fat and sugar reverses anxiety-like behaviour induced by limited nesting in male rats: Impacts on hippocampal markers. Psychoneuroendocrinology 2016; 68:202-9. [PMID: 26999723 DOI: 10.1016/j.psyneuen.2016.03.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 02/01/2016] [Accepted: 03/09/2016] [Indexed: 12/29/2022]
Abstract
Stress exposure during early development is known to produce long-term mental health deficits. Stress promotes poor lifestyle choices such as poor diet. Early life adversity and diets high in fat and sugar (HFHS) are known to affect anxiety and memory. However additive effects of HFHS and stress during early development are less explored. Here, we examined whether early life stress (ELS) simulated by limited nesting (LN) induces anxiety-like behaviour and cognitive deficits that are modulated by HFHS diet. We examined key hippocampal markers involved in anxiety and cognition, testing the hypothesis that post-weaning HFHS following ELS would ameliorate anxiety-like behaviour but worsen memory and associated hippocampal changes. Sprague-Dawley rats were exposed to LN, postnatal days 2-9, and at weaning, male siblings were given unlimited access to chow or HFHS resulting in (Con-Chow, Con-HFHS, LN-Chow, LN-HFHS, n=11-15/group). Anxiety-like behaviour was assessed by Elevated Plus Maze (EPM) at 10 weeks and spatial and object recognition tested at 11 weeks of age. Rats were culled at 13 weeks. Hippocampal mRNA expression was measured using TaqMan(®) Array Micro Fluidic cards (Life Technologies). As expected HFHS diet increased body weight; LN and control rats had similar weights at 13 weeks, energy intake was also similar across groups. LN-Chow rats showed increased anxiety-like behaviour relative to control rats, but this was reversed by HFHS diet. Spatial and object recognition memory were unaltered by LN exposure or consumption of HFHS diet. Hippocampal glucocorticoid receptor (GR) protein was not affected by LN exposure in chow rats, but was increased by 45% in HFHS rats relative to controls. Hippocampal genes involved in plasticity and mood regulation, GSKα and GSKβ were affected, with reductions in GSKβ under both diet conditions, and reduced GSKα only in LN-HFHS versus Con-HFHS. Interestingly, HFHS diet and LN exposure independently reduced expression of Akt3 mRNA, a key gene involved post-natal brain development. In summary, while an energy rich diet ameliorated anxiety-like behaviour induced by LN exposure, it significantly altered key genes that are essential for hippocampal development.
Collapse
Affiliation(s)
- Jayanthi Maniam
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney 2052, New South Wales, Australia
| | - Christopher P Antoniadis
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney 2052, New South Wales, Australia
| | - Vivian Le
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney 2052, New South Wales, Australia
| | - Margaret J Morris
- Department of Pharmacology, School of Medical Sciences, UNSW Australia, Sydney 2052, New South Wales, Australia.
| |
Collapse
|
188
|
Arp JM, Ter Horst JP, Loi M, den Blaauwen J, Bangert E, Fernández G, Joëls M, Oitzl MS, Krugers HJ. Blocking glucocorticoid receptors at adolescent age prevents enhanced freezing between repeated cue-exposures after conditioned fear in adult mice raised under chronic early life stress. Neurobiol Learn Mem 2016; 133:30-38. [PMID: 27246249 DOI: 10.1016/j.nlm.2016.05.009] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 04/19/2016] [Accepted: 05/27/2016] [Indexed: 12/11/2022]
Abstract
Early life adversity can have long-lasting impact on learning and memory processes and increase the risk to develop stress-related psychopathologies later in life. In this study we investigated (i) how chronic early life stress (ELS) - elicited by limited nesting and bedding material from postnatal day 2 to 9 - affects conditioned fear in adult mice and (ii) whether these effects can be prevented by blocking glucocorticoid receptors (GRs) at adolescent age. In adult male and female mice, ELS did not affect freezing behavior to the first tone 24h after training in an auditory fear-conditioning paradigm. Exposure to repeated tones 24h after training also resulted in comparable freezing behavior in ELS and control mice, both in males and females. However, male (but not female) ELS compared to control mice showed significantly more freezing behavior between the tone-exposures, i.e. during the cue-off periods. Intraperitoneal administration of the GR antagonist RU38486 during adolescence (on postnatal days 28-30) fully prevented enhanced freezing behavior during the cue-off period in adult ELS males. Western blot analysis revealed no effects of ELS on hippocampal expression of glucocorticoid receptors, neither at postnatal day 28 nor at adult age, when mice were behaviorally tested. We conclude that ELS enhances freezing behavior in adult mice in a potentially safe context after cue-exposure, which can be normalized by brief blockade of glucocorticoid receptors during the critical developmental window of adolescence.
Collapse
Affiliation(s)
- J Marit Arp
- SILS-Center for Neuroscience, University of Amsterdam, The Netherlands
| | | | - Manila Loi
- Dept. Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands
| | - Jan den Blaauwen
- SILS-Center for Neuroscience, University of Amsterdam, The Netherlands
| | - Eline Bangert
- SILS-Center for Neuroscience, University of Amsterdam, The Netherlands
| | - Guillén Fernández
- Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marian Joëls
- Dept. Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, The Netherlands
| | - Melly S Oitzl
- SILS-Center for Neuroscience, University of Amsterdam, The Netherlands
| | - Harm J Krugers
- SILS-Center for Neuroscience, University of Amsterdam, The Netherlands.
| |
Collapse
|
189
|
Won H, Abdul Manaf Z, Mat Ludin AF, Shahar S. Wide range of body composition measures are associated with cognitive function in community-dwelling older adults. Geriatr Gerontol Int 2016; 17:554-560. [PMID: 27231255 DOI: 10.1111/ggi.12753] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 11/23/2015] [Accepted: 01/04/2016] [Indexed: 12/29/2022]
Abstract
AIM Studies of the association between body composition, both body fat and body muscle, and cognitive function are rarely reported. The aim of the present study was to determine the association between a wide range of body composition measures with cognitive function in older adults. METHODS A total of 2322 Malaysian older adults aged 60 years and older were recruited using multistage random sampling in a population-based cross-sectional study. Out of 2322 older adults recruited, 2309 (48% men) completed assessments on cognitive function and body composition. Cognitive functions were assessed using the Malay version of the Mini-Mental State Examination, the Bahasa Malaysia version of Montreal Cognitive Assessment, Digit Span Test, Digit Symbol Test and Rey Auditory Verbal Learning Test. Body composition included body mass index, mid-upper arm circumference, waist circumference, calf circumference, waist-to-hip ratio, percentage body fat and skeletal muscle mass. RESULTS The association between body composition and cognitive functions was analyzed using multiple linear regression. After adjustment for age, education years, hypertension, hypercholesterolemia, diabetes mellitus, depression, smoking status and alcohol consumption, we found that calf circumference appeared as a significant predictor for all cognitive tests among both men and women (P < 0.05), except for the Rey Auditory Verbal Learning Test. Waist-to-hip ratio was detected as a significant predictor for all cognitive tests among women (P < 0.05), but was only a significant predictor for the Bahasa Malaysia version of Montreal Cognitive Assessment among men (P < 0.05). CONCLUSIONS These results suggest that there is a need to maintain muscle mass and lower adipose tissue among older adults for optimal cognitive function. Geriatr Gerontol Int 2017; 17: 554-560.
Collapse
Affiliation(s)
- Huiloo Won
- Nutrition Program, Faculty of Health Sciences, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Zahara Abdul Manaf
- Dietetics Program, Faculty of Health Sciences, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Arimi Fitri Mat Ludin
- Biomedical Science Program, School of Diagnostic and Applied Health Sciences, Faculty of Health Sciences, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Suzana Shahar
- Dietetics Program, Faculty of Health Sciences, University Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
190
|
Hsiao YM, Tsai TC, Lin YT, Chen CC, Huang CC, Hsu KS. Early life stress dampens stress responsiveness in adolescence: Evaluation of neuroendocrine reactivity and coping behavior. Psychoneuroendocrinology 2016; 67:86-99. [PMID: 26881834 DOI: 10.1016/j.psyneuen.2016.02.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/03/2016] [Accepted: 02/05/2016] [Indexed: 02/06/2023]
Abstract
Stressful experiences during early life (ELS) can affect brain development, thereby exerting a profound and long-lasting influence on mental development and psychological health. The stress inoculation hypothesis presupposes that individuals who have early experienced an attenuated form of stressors may gain immunity to its more virulent forms later in life. Increasing evidence demonstrates that ELS may promote the development of subsequent stress resistance, but the mechanisms underlying such adaptive changes are not fully understood. The present study evaluated the impact of fragmented dam-pup interactions by limiting the bedding and nesting material in the cage during postnatal days 2-9, a naturalistic animal model of chronic ELS, on the physiological and behavioral responses to different stressors in adolescent mice and characterized the possible underlying mechanisms. We found that ELS mice showed less social interaction deficits after chronic social defeat stress and acute restraint-tailshock stress-induced impaired long-term potentiation (LTP) and enhanced long-term depression (LTD) in hippocampal CA1 region compared with control mice. The effects of ELS on LTP and LTD were rescued by adrenalectomy. While ELS did not cause alterations in basal emotional behaviors, it significantly enhanced stress coping behaviors in both the tail suspension and the forced swimming tests. ELS mice exhibited a significant decrease in corticosterone response and trafficking of glucocorticoid receptors to the nucleus in response to acute restraint stress. Altogether, our data support the hypothesis that stress inoculation training, via early exposure to manageable stress, may enhance resistance to other unrelated extreme stressors in adolescence.
Collapse
Affiliation(s)
- Young-Ming Hsiao
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tsung-Chih Tsai
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Ting Lin
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Chung Chen
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chiung-Chun Huang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuei-Sen Hsu
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
191
|
Lv YL, Wu ZZ, Chen LX, Wu BX, Chen LL, Qin GC, Gui B, Zhou JY. Neuroprotective effects of tetrandrine against vascular dementia. Neural Regen Res 2016; 11:454-9. [PMID: 27127485 PMCID: PMC4829011 DOI: 10.4103/1673-5374.179058] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Tetrandrine is one of the major active ingredients in Menispermaceae Stephania tetrandra S. Moore, and has specific therapeutic effects in ischemic cerebrovascular disease. Its use in vascular dementia has not been studied fully. Here, we investigated whether tetrandrine would improve behavioral and cellular impairments in a two-vessel occlusion rat model of chronic vascular dementia. Eight weeks after model establishment, rats were injected intraperitoneally with 10 or 30 mg/kg tetrandrine every other day for 4 weeks. Behavioral assessment in the Morris water maze showed that model rats had longer escape latencies in training trials, and spent less time swimming in the target quadrant in probe trials, than sham-operated rats. However, rats that had received tetrandrine showed shorter escape latencies and longer target quadrant swimming time than untreated model rats. Hematoxylin-eosin and Nissl staining revealed less neuronal necrosis and pathological damage, and more living cells, in the hippocampus of rats treated with tetrandrine than in untreated model rats. Western blot assay showed that interleukin-1β expression, and phosphorylation of the N-methyl-D-aspartate 2B receptor at tyrosine 1472, were lower in model rats that received tetrandrine than in those that did not. The present findings suggest that tetrandrine may be neuroprotective in chronic vascular dementia by reducing interleukin-1β expression, N-methyl-D-aspartate receptor 2B phosphorylation at tyrosine 1472, and neuronal necrosis.
Collapse
Affiliation(s)
- Yan-Ling Lv
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ze-Zhi Wu
- Key Laboratory of Biorheological Science and Technology of the State Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Li-Xue Chen
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bai-Xue Wu
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lian-Lian Chen
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guang-Cheng Qin
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bei Gui
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ji-Ying Zhou
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
192
|
Amos-Kroohs RM, Graham DL, Grace CE, Braun AA, Schaefer TL, Skelton MR, Vorhees CV, Williams MT. Developmental stress and lead (Pb): Effects of maternal separation and/or Pb on corticosterone, monoamines, and blood Pb in rats. Neurotoxicology 2016; 54:22-33. [PMID: 26943976 DOI: 10.1016/j.neuro.2016.02.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 02/17/2016] [Accepted: 02/28/2016] [Indexed: 01/13/2023]
Abstract
The level of lead (Pb) exposure in children has decreased dramatically since restrictions on its use were implemented. However, even with restrictions, children are exposed to Pb and still present with cognitive and behavioral deficits. One prominent aspect of the exposome of these children is that many come from low social economic status (SES) conditions, and low SES is associated with stress. In order to compare the combined effects of early stress and Pb, Sprague-Dawley rats were exposed to vehicle or Pb either alone or in combination with maternal separation stress during brain development (i.e., postnatal day (P)4-P11, P19, or P28). Maternally separated/isolated pups had lower body and thymus weights during exposure and had increased levels of blood Pb compared with vehicle controls. Isolation, but not Pb, affected the response to an acute stressor (standing in shallow water) when assessed on P19 and P29, but not earlier on P11. Interactions of Pb and isolation were found on monoamines in the neostriatum, hippocampus, and hypothalamus on turnover but not on levels, and most changes were on dopamine turnover. Isolation had greater short-term effects than Pb. Interactions were dependent on age, sex, and acute stress.
Collapse
Affiliation(s)
- Robyn M Amos-Kroohs
- Department of Nutritional Sciences, University of Wisconsin-Madison, 1415 Linden Dr., Madison, WI 53706, United States.
| | - Devon L Graham
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, United States; University of Cincinnati College of Medicine, Cincinnati, OH 45229, United States.
| | - Curtis E Grace
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, United States; University of Cincinnati College of Medicine, Cincinnati, OH 45229, United States.
| | - Amanda A Braun
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, United States; University of Cincinnati College of Medicine, Cincinnati, OH 45229, United States.
| | - Tori L Schaefer
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, United States; University of Cincinnati College of Medicine, Cincinnati, OH 45229, United States.
| | - Matthew R Skelton
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, United States; University of Cincinnati College of Medicine, Cincinnati, OH 45229, United States.
| | - Charles V Vorhees
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, United States; University of Cincinnati College of Medicine, Cincinnati, OH 45229, United States.
| | - Michael T Williams
- Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH, United States; University of Cincinnati College of Medicine, Cincinnati, OH 45229, United States.
| |
Collapse
|
193
|
Koe AS, Ashokan A, Mitra R. Short environmental enrichment in adulthood reverses anxiety and basolateral amygdala hypertrophy induced by maternal separation. Transl Psychiatry 2016; 6:e729. [PMID: 26836417 PMCID: PMC4872421 DOI: 10.1038/tp.2015.217] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 10/18/2015] [Accepted: 12/02/2015] [Indexed: 12/22/2022] Open
Abstract
Maternal separation during early childhood results in greater sensitivity to stressors later in adult life. This is reflected as greater propensity to develop stress-related disorders in humans and animal models, including anxiety and depression. Environmental enrichment (EE) reverses some of the damaging effects of maternal separation in rodent models when provided during peripubescent life, temporally proximal to the separation. It is presently unknown if EE provided outside this critical window can still rescue separation-induced anxiety and neural plasticity. In this report we use a rat model to demonstrate that a single short episode of EE in adulthood reduced anxiety-like behaviour in maternally separated rats. We further show that maternal separation resulted in hypertrophy of dendrites and increase in spine density of basolateral amygdala neurons in adulthood, long after initial stress treatment. This is congruent with prior observations showing centrality of basolateral amygdala hypertrophy in anxiety induced by stress during adulthood. In line with the ability of the adult enrichment to rescue stress-induced anxiety, we show that enrichment renormalized stress-induced structural expansion of the amygdala neurons. These observations argue that behavioural plasticity induced by early adversity can be rescued by environmental interventions much later in life, likely mediated by ameliorating effects of enrichment on basolateral amygdala plasticity.
Collapse
Affiliation(s)
- A S Koe
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - A Ashokan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - R Mitra
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore,School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore. E-mail:
| |
Collapse
|
194
|
Kanatsou S, Ter Horst JP, Harris AP, Seckl JR, Krugers HJ, Joëls M. Effects of Mineralocorticoid Receptor Overexpression on Anxiety and Memory after Early Life Stress in Female Mice. Front Behav Neurosci 2016; 9:374. [PMID: 26858618 PMCID: PMC4726803 DOI: 10.3389/fnbeh.2015.00374] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 12/30/2015] [Indexed: 11/29/2022] Open
Abstract
Early-life stress (ELS) is a risk factor for the development of psychopathology, particularly in women. Human studies have shown that certain haplotypes of NR3C2, encoding the mineralocorticoid receptor (MR), that result in gain of function, may protect against the consequences of stress exposure, including childhood trauma. Here, we tested the hypothesis that forebrain-specific overexpression of MR in female mice would ameliorate the effects of ELS on anxiety and memory in adulthood. We found that ELS increased anxiety, did not alter spatial discrimination and reduced contextual fear memory in adult female mice. Transgenic overexpression of MR did not alter anxiety but affected spatial memory performance and enhanced contextual fear memory formation. The effects of ELS on anxiety and contextual fear were not affected by transgenic overexpression of MR. Thus, MR overexpression in the forebrain does not represent a major resilience factor to early life adversity in female mice.
Collapse
Affiliation(s)
- Sofia Kanatsou
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center UtrechtUtrecht, Netherlands; Swammerdam Institute for Life Sciences, Center for Neuroscience, University of AmsterdamAmsterdam, Netherlands
| | - Judith P Ter Horst
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam Amsterdam, Netherlands
| | - Anjanette P Harris
- Endocrinology Unit, Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh Edinburgh, UK
| | - Jonathan R Seckl
- Endocrinology Unit, Centre for Cardiovascular Science, Queen's Medical Research Institute, The University of Edinburgh Edinburgh, UK
| | - Harmen J Krugers
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam Amsterdam, Netherlands
| | - Marian Joëls
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht Utrecht, Netherlands
| |
Collapse
|
195
|
Tochitani S, Ikeno T, Ito T, Sakurai A, Yamauchi T, Matsuzaki H. Administration of Non-Absorbable Antibiotics to Pregnant Mice to Perturb the Maternal Gut Microbiota Is Associated with Alterations in Offspring Behavior. PLoS One 2016; 11:e0138293. [PMID: 26789865 PMCID: PMC4720425 DOI: 10.1371/journal.pone.0138293] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 12/03/2015] [Indexed: 12/13/2022] Open
Abstract
There is increasing evidence that the gut microbiota plays a major role in host health and disease. In this study, we examined whether perturbation of the maternal gut microbiota during pregnancy, induced by administration of non-absorbable antibiotics to pregnant dams, influences the behavior of offspring. Terminal restriction fragment length polymorphism analyses of fecal bacterial composition showed that the relative abundance of the bacterial order Lactobacillales was lower in offspring born from antibiotic-treated dams (20.7±3.4%) than in control offspring (42.1±6.2%) at P24, while the relative abundance of the bacterial family Clostridium subcluster XIVa was higher in offspring born from antibiotic-treated dams (34.2±5.0%) than in control offspring (16.4±3.3%). Offspring born from antibiotic-treated dams exhibited low locomotor activity in both familiar and novel environments, and preferred to explore in the peripheral area of an unfamiliar field at postnatal week 4. At postnatal weeks 7–8, no difference was observed in the level of locomotor activity between control offspring and offspring from antibiotic-treated dams, while the tendency for the offspring from antibiotic-treated dams to be less engaged in exploring the inside area was still observed. The behavioral phenotypes of the offspring from antibiotic-treated dams at postnatal week 4 could be rescued to a considerable extent through fostering of these offspring by normal dams from postnatal day 1. Although the detailed underlying mechanisms are not fully elucidated, the present results suggest that administration of non-absorbable antibiotics to pregnant dams to perturb the maternal gut microbiota during pregnancy leads to alterations in the behavior of their offspring.
Collapse
Affiliation(s)
- Shiro Tochitani
- Division of Development of Mental Functions, Research Center for Child Mental Development, University of Fukui, Fukui 910–1193, Japan
- Division of Developmental Higher Brain Functions, Department of Child Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Osaka 565–0871, Japan
- Research and Education Program for Life Science, University of Fukui, Fukui 910–8507, Japan
- * E-mail:
| | - Takahiro Ikeno
- School of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui 910–1193, Japan
| | - Tatsuhito Ito
- School of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui 910–1193, Japan
| | - Asuka Sakurai
- School of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui 910–1193, Japan
| | - Tomoki Yamauchi
- School of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui 910–1193, Japan
| | - Hideo Matsuzaki
- Division of Development of Mental Functions, Research Center for Child Mental Development, University of Fukui, Fukui 910–1193, Japan
- Division of Developmental Higher Brain Functions, Department of Child Development, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of Fukui, Osaka 565–0871, Japan
- Research and Education Program for Life Science, University of Fukui, Fukui 910–8507, Japan
- School of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui 910–1193, Japan
| |
Collapse
|
196
|
Liu R, Yang XD, Liao XM, Xie XM, Su YA, Li JT, Wang XD, Si TM. Early postnatal stress suppresses the developmental trajectory of hippocampal pyramidal neurons: the role of CRHR1. Brain Struct Funct 2016; 221:4525-4536. [DOI: 10.1007/s00429-016-1182-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 01/05/2016] [Indexed: 11/29/2022]
|
197
|
Chen Y, Baram TZ. Toward Understanding How Early-Life Stress Reprograms Cognitive and Emotional Brain Networks. Neuropsychopharmacology 2016; 41:197-206. [PMID: 26105143 PMCID: PMC4677123 DOI: 10.1038/npp.2015.181] [Citation(s) in RCA: 308] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 05/26/2015] [Accepted: 06/19/2015] [Indexed: 01/19/2023]
Abstract
Vulnerability to emotional disorders including depression derives from interactions between genes and environment, especially during sensitive developmental periods. Adverse early-life experiences provoke the release and modify the expression of several stress mediators and neurotransmitters within specific brain regions. The interaction of these mediators with developing neurons and neuronal networks may lead to long-lasting structural and functional alterations associated with cognitive and emotional consequences. Although a vast body of work has linked quantitative and qualitative aspects of stress to adolescent and adult outcomes, a number of questions are unclear. What distinguishes 'normal' from pathologic or toxic stress? How are the effects of stress transformed into structural and functional changes in individual neurons and neuronal networks? Which ones are affected? We review these questions in the context of established and emerging studies. We introduce a novel concept regarding the origin of toxic early-life stress, stating that it may derive from specific patterns of environmental signals, especially those derived from the mother or caretaker. Fragmented and unpredictable patterns of maternal care behaviors induce a profound chronic stress. The aberrant patterns and rhythms of early-life sensory input might also directly and adversely influence the maturation of cognitive and emotional brain circuits, in analogy to visual and auditory brain systems. Thus, unpredictable, stress-provoking early-life experiences may influence adolescent cognitive and emotional outcomes by disrupting the maturation of the underlying brain networks. Comprehensive approaches and multiple levels of analysis are required to probe the protean consequences of early-life adversity on the developing brain. These involve integrated human and animal-model studies, and approaches ranging from in vivo imaging to novel neuroanatomical, molecular, epigenomic, and computational methodologies. Because early-life adversity is a powerful determinant of subsequent vulnerabilities to emotional and cognitive pathologies, understanding the underlying processes will have profound implications for the world's current and future children.
Collapse
Affiliation(s)
- Yuncai Chen
- Department of Pediatrics, University of California, Irvine, CA, USA
| | - Tallie Z Baram
- Department of Pediatrics, University of California, Irvine, CA, USA
- Department of Anatomy/Neurobiology, University of California, Irvine, CA, USA
- Department of Neurology, University of California, Irvine, CA, USA
| |
Collapse
|
198
|
Molet J, Heins K, Zhuo X, Mei YT, Regev L, Baram TZ, Stern H. Fragmentation and high entropy of neonatal experience predict adolescent emotional outcome. Transl Psychiatry 2016; 6:e702. [PMID: 26731439 PMCID: PMC5068874 DOI: 10.1038/tp.2015.200] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/30/2015] [Accepted: 11/05/2015] [Indexed: 01/22/2023] Open
Abstract
Vulnerability to emotional disorders including depression derives from interactions between genes and environment, especially during sensitive developmental periods. Across evolution, maternal care is a key source of environmental sensory signals to the developing brain, and a vast body of work has linked quantitative and qualitative aspects of maternal care to emotional outcome in children and animals. However, the fundamental properties of maternal signals, that promote advantageous vs pathological outcomes in the offspring, are unknown and have been a topic of intense study. We studied emotional outcomes of adolescent rats reared under routine or impoverished environments, and used mathematical approaches to analyze the nurturing behaviors of the dams. Unexpectedly, whereas the quantity and typical qualities of maternal care behaviors were indistinguishable in the two environments, their patterns and rhythms differed drastically and influenced emotional outcomes. Specifically, unpredictable, fragmented maternal care patterns translated into high-entropy rates of sensory signals to the offspring in the impoverished cages. During adolescence, these offspring had significant reductions in sucrose preference and in peer-play, two independent measures of the ability to experience pleasure. This adolescent anhedonia, often a harbinger of later depression, was not accompanied by measures of anxiety or helplessness. Dopaminergic pleasure circuits underlying anhedonia are engaged by predictable sequences of events, and predictable sensory signals during neonatal periods may be critical for their maturation. Conversely, unpredictability maternal-derived signals may disrupt these developmental processes, provoking anhedonia. In sum, high-entropy and fragmented patterns of maternal-derived sensory input to the developing brain predicts, and might promote, the development of anhedonia in rodents, with potential clinical implications.
Collapse
Affiliation(s)
- J Molet
- Department of Anatomy and Neurobiology, University of California-Irvine, Irvine, CA, USA
| | - K Heins
- Department of Statistics, Donald Bren School of Information and Computer Sciences, University of California-Irvine, Irvine, CA, USA
| | - X Zhuo
- Department of Anatomy and Neurobiology, University of California-Irvine, Irvine, CA, USA
| | - Y T Mei
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
| | - L Regev
- Department of Anatomy and Neurobiology, University of California-Irvine, Irvine, CA, USA
| | - T Z Baram
- Department of Anatomy and Neurobiology, University of California-Irvine, Irvine, CA, USA,Department of Pediatrics, University of California-Irvine, Irvine, CA, USA,Department of Neurology, School of Medicine, University of California-Irvine, Irvine, CA, USA,Departments of Pediatrics; Anatomy/Neurobiology; Neurology, University of California-Irvine, Medical Sciences I, ZOT: 4475, Irvine, CA 92697-4475, USA. E-mail:
| | - H Stern
- Department of Statistics, Donald Bren School of Information and Computer Sciences, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
199
|
Moreno Gudiño H, Carías Picón D, de Brugada Sauras I. Dietary choline during periadolescence attenuates cognitive damage caused by neonatal maternal separation in male rats. Nutr Neurosci 2015; 20:327-335. [DOI: 10.1080/1028415x.2015.1126444] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Hayarelis Moreno Gudiño
- Department of Experimental Psychology and Physiology of behavior, University of Granada, Spain
- Department of Biological and Biochemical Processes, Simón Bolívar University, Caracas, Venezuela
| | - Diamela Carías Picón
- Department of Biological and Biochemical Processes, Simón Bolívar University, Caracas, Venezuela
| | | |
Collapse
|
200
|
McFalls AJ, Imperio CG, Bixler G, Freeman WM, Grigson PS, Vrana KE. Reward devaluation and heroin escalation is associated with differential expression of CRF signaling genes. Brain Res Bull 2015; 123:81-93. [PMID: 26655889 DOI: 10.1016/j.brainresbull.2015.11.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/04/2015] [Accepted: 11/05/2015] [Indexed: 01/08/2023]
Abstract
One of the most damaging aspects of drug addiction is the degree to which natural rewards (family, friends, employment) are devalued in favor of seeking, obtaining and taking drugs. We have utilized an animal model of reward devaluation and heroin self-administration to explore the role of the coricotropin releasing factor (CRF) pathway. Given access to a saccharin cue followed by the opportunity to self-administer heroin, animals will parse into distinct phenotypes that suppress their saccharin intake (in favor of escalating heroin self-administration) or vice versa. We find that large saccharin suppressors (large heroin takers) demonstrate increased mRNA expression for elements of the CRF signaling pathway (CRF, CRF receptors and CRF binding protein) within the hippocampus, medial prefrontal cortex and the ventral tegmental area. Moreover, there were no gene expression changes of these components in the nucleus accumbens. Use of bisulfite conversion sequencing suggests that changes in CRF binding protein and CRF receptor gene expression may be mediated by differential promoter methylation.
Collapse
Affiliation(s)
- Ashley J McFalls
- Departments of Pharmacology, Penn State College of Medicine, Hershey, PA, United States
| | - Caesar G Imperio
- Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States
| | - Georgina Bixler
- Departments of Pharmacology, Penn State College of Medicine, Hershey, PA, United States
| | - Willard M Freeman
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Patricia Sue Grigson
- Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA, United States
| | - Kent E Vrana
- Departments of Pharmacology, Penn State College of Medicine, Hershey, PA, United States.
| |
Collapse
|