1
|
Rosenkranz JA. Shaping behaviors through social experience and their proposed sensitivity to stress. Learn Mem 2024; 31:a053926. [PMID: 39681461 DOI: 10.1101/lm.053926.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/09/2024] [Indexed: 12/18/2024]
Abstract
Mammals have evolved with a range of innate drives, such as thirst and hunger, that promote motivated behaviors to ensure survival. A drive for social engagement promotes social interaction and bond formation. While a stable social environment maintains the opportunity for resource sharing and protection, an additional benefit is provided by the social transmission of information. Social experiences, and information obtained from conspecifics, can be used to learn about threats and opportunities in the environment. This review examines the primary forms of social learning and how they can shape behavior. Additionally, while there is much known about the effects of stress on learning and memory, there is much less known about its effects on social learning and memory. This review will therefore dissect the major factors that contribute to social learning and propose how stress may impact these factors. This may serve as a way to formulate new hypotheses about how stress might impact social learning and the effects of social experiences on behavior.
Collapse
Affiliation(s)
- J Amiel Rosenkranz
- Center for Neurobiology of Stress Resilience and Psychiatric Disorders, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064, USA
| |
Collapse
|
2
|
Ahmed S, Polis B, Jamwal S, Sanganahalli BG, MacDowell Kaswan Z, Islam R, Kim D, Bowers C, Giuliano L, Biederer T, Hyder F, Kaffman A. Transient impairment in microglial function causes sex-specific deficits in synaptic maturity and hippocampal function in mice exposed to early adversity. Brain Behav Immun 2024; 122:95-109. [PMID: 39134183 PMCID: PMC11402597 DOI: 10.1016/j.bbi.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024] Open
Abstract
Abnormal development and function of the hippocampus are two of the most consistent findings in humans and rodents exposed to early-life adversity (ELA), with males often being more affected than females. Using the limited bedding (LB) paradigm as a rodent model of ELA, we found that male adolescent mice that had been exposed to LB exhibit significant deficits in contextual fear conditioning and synaptic connectivity in the hippocampus, which are not observed in females. This is linked to altered developmental refinement of connectivity, with LB severely impairing microglial-mediated synaptic pruning in the hippocampus of male and female pups on postnatal day 17 (P17), but not in adolescent P33 mice when levels of synaptic engulfment by microglia are substantially lower. Since the rodent hippocampus undergoes intense synaptic pruning during the second and third weeks of life, we investigated whether microglia are required for the synaptic and behavioral aberrations observed in adolescent LB mice. Indeed, transient ablation of microglia from P13-21 in normally developing mice caused sex-specific behavioral and synaptic abnormalities similar to those observed in adolescent LB mice. Furthermore, chemogenetic activation of microglia during the same period reversed the microglial-mediated phagocytic deficits at P17 and restored normal contextual fear conditioning and synaptic connectivity in adolescent LB male mice. Our data support an additional contribution of astrocytes in the sex-specific effects of LB, with increased expression of the membrane receptor MEGF10 and enhanced synaptic engulfment in hippocampal astrocytes of 17-day-old LB females, but not in LB male littermates. These findings suggest a potential compensatory mechanism that may explain the relative resilience of LB females. Collectively, our study highlights a novel role for glial cells in mediating sex-specific hippocampal deficits in a mouse model of ELA.
Collapse
Affiliation(s)
- Sahabuddin Ahmed
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Baruh Polis
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Sumit Jamwal
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Basavaraju G Sanganahalli
- Department of Radiology & Biomedical Imaging and Magnetic Resonance Research Center, Yale University, New Haven, CT, 06520, USA
| | - Zoe MacDowell Kaswan
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Rafiad Islam
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Dana Kim
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Christian Bowers
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Lauryn Giuliano
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Thomas Biederer
- Department of Neurology, Yale School of Medicine, 100 College Street, New Haven, CT 06510, USA
| | - Fahmeed Hyder
- Department of Radiology & Biomedical Imaging and Magnetic Resonance Research Center, Yale University, New Haven, CT, 06520, USA; Department of Biomedical Engineering, Yale University, New Haven, CT, 06519, USA
| | - Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA.
| |
Collapse
|
3
|
Hope SF, Willgohs KR, Dittakul S, Plotnik JM. Do elephants really never forget? What we know about elephant memory and a call for further investigation. Learn Behav 2024:10.3758/s13420-024-00655-y. [PMID: 39438402 DOI: 10.3758/s13420-024-00655-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2024] [Indexed: 10/25/2024]
Abstract
Despite popular culture's promotion of the elephant's ability to "never forget," there is remarkably limited empirical research on the memory capacities of any living elephant species (Asian, Elephas maximus; African savanna, Loxodonta africana; African forest, Loxodonta cyclotis). A growing body of literature on elephant cognition and behavioral ecology has provided insight into the elephant's ability to behave flexibly in changing physical and social environments, but little direct evidence of how memory might relate to this flexibility exists. In this paper, we review and discuss the potential relationships between what we know about elephant cognition and behavior and the elephants' memory for the world around them as they navigate their physical, social, and spatial environments. We also discuss future directions for investigating elephant memory and implications for such research on elephant conservation and human-elephant conflict mitigation.
Collapse
Affiliation(s)
- Sydney F Hope
- Department of Psychology, Hunter College, City University of New York, 695 Park Avenue, New York, NY, 10065, USA.
| | - Kaitlyn R Willgohs
- Department of Psychology, Hunter College, City University of New York, 695 Park Avenue, New York, NY, 10065, USA
- Department of Psychology, The Graduate Center, City University of New York, New York, NY, 10016, USA
| | - Sangpa Dittakul
- Department of Psychology, Hunter College, City University of New York, 695 Park Avenue, New York, NY, 10065, USA
- Golden Triangle Asian Elephant Foundation, Chiang Saen, Chiang Rai, 57150, Thailand
| | - Joshua M Plotnik
- Department of Psychology, Hunter College, City University of New York, 695 Park Avenue, New York, NY, 10065, USA.
- Department of Psychology, The Graduate Center, City University of New York, New York, NY, 10016, USA.
| |
Collapse
|
4
|
Jin J, Fu C, Xia J, Luo H, Wang X, Chen S, Mao H, Yuan K, Lu L, Xiong W, Zou G. Cannabidiol ameliorates cognitive decline in 5×FAD mouse model of Alzheimer's disease through potentiating the function of extrasynaptic glycine receptors. Mol Psychiatry 2024:10.1038/s41380-024-02789-x. [PMID: 39396064 DOI: 10.1038/s41380-024-02789-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/14/2024]
Abstract
Emerging evidence supports the therapeutic potential of cannabinoids in Alzheimer's disease (AD), but the underlying mechanism upon how cannabinoids impact brain cognition and AD pathology remains unclear. Here we show that chronic cannabidiol (CBD) administration significantly mitigates cognitive deficiency and hippocampal β-amyloid (Aβ) pathology in 5×FAD mouse model of AD. CBD achieves its curative effect mainly through potentiating the function of inhibitory extrasynaptic glycine receptor (GlyR) in hippocampal dentate gyrus (DG). Based on the in vitro and in vivo electrophysiological recording and calcium imaging, CBD mediated anti-AD effects via GlyR are mainly accomplished by decreasing neuronal hyperactivity of granule cells in the DG of AD mice. Furthermore, the AAV-mediated ablation of DG GlyRα1, or the GlyRα1S296A mutation that exclusively disrupts CBD binding, significantly intercepts the anti-AD effect of CBD. These findings suggest a GlyR dependent mechanism underlying the therapeutic potential of CBD in the treatment of AD.
Collapse
Grants
- 32225020, 91849206, 91942315, 92049304, 32121002, 81901157, 82241032 National Natural Science Foundation of China (National Science Foundation of China)
- 32225020, 91849206, 91942315, 92049304, 32121002 National Natural Science Foundation of China (National Science Foundation of China)
Collapse
Affiliation(s)
- Jin Jin
- Department of neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Chonglei Fu
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan, 250117, China
| | - Jing Xia
- Department of neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Heyi Luo
- Department of neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Xianglian Wang
- Department of neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Si Chen
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Huanhuan Mao
- Department of neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Kai Yuan
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Chinese Academy of Medical Sciences Research Unit (No.2018RU006), Peking University, 100191, Beijing, China
| | - Lin Lu
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan, 250117, China.
- Peking University Sixth Hospital, Peking University Institute of Mental Health, NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Chinese Academy of Medical Sciences Research Unit (No.2018RU006), Peking University, 100191, Beijing, China.
| | - Wei Xiong
- Department of neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
- Anhui Province Key Laboratory of Biomedical Imaging and Intelligent Processing, Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, 230088, China.
- CAS Key Laboratory of Brain Function and Disease, Hefei, 230026, China.
- Anhui Province Key Laboratory of Biomedical Aging Research, Hefei, 230026, China.
| | - Guichang Zou
- Institute of Brain Science and Brain-inspired Research, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
- Shandong Institute of Brain Science and Brain-inspired Research, Jinan, 250117, China.
| |
Collapse
|
5
|
Mahmoudian M, Lorigooini Z, Rahimi-Madiseh M, Shabani S, Amini-Khoei H. Protective effects of rosmarinic acid against autistic-like behaviors in a mouse model of maternal separation stress: behavioral and molecular amendments. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7819-7828. [PMID: 38730077 DOI: 10.1007/s00210-024-03143-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 05/02/2024] [Indexed: 05/12/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder with worldwide increasing incidence. Maternal separation (MS) stress at the beginning of life with its own neuroendocrine changes can provide the basis for development of ASD. Rosmarinic acid (RA) is a phenolic compound with a protective effect in neurodegenerative diseases. The aim of this study was to determine the effect of RA on autistic-like behaviors in maternally separated mice focusing on its possible effects on neuroimmune response and nitrite levels in the hippocampus. In this study, 40 mice were randomly divided into five groups of control (received normal saline (1 ml/kg)) and MS that were treated with normal saline (1 ml/kg) or doses of 1, 2, and 4 mg/kg RA, respectively, for 14 days. Three-chamber sociability, shuttle box, and marble burying tests were used to investigate autistic-like behaviors. Nitrite level and gene expression of inflammatory cytokines including TNF-α, IL-1β, TLR4, and iNOS were assessed in the hippocampus. The results showed that RA significantly increased the social preference and social novelty indexes, as well as attenuated impaired passive avoidance memory and the occurrence of repetitive and obsessive behaviors in the MS mice. RA reduced the nitrite level and gene expression of inflammatory cytokines in the hippocampus. RA, probably via attenuation of the nitrite level as well as of the neuroimmune response in the hippocampus, mitigated autistic-like behaviors in maternally separated mice.
Collapse
Affiliation(s)
- Maziar Mahmoudian
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Lorigooini
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Rahimi-Madiseh
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sahreh Shabani
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hossein Amini-Khoei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
6
|
Calabrese V, Osakabe N, Siracusa R, Modafferi S, Di Paola R, Cuzzocrea S, Jacob UM, Fritsch T, Abdelhameed AS, Rashan L, Wenzel U, Franceschi C, Calabrese EJ. Transgenerational hormesis in healthy aging and antiaging medicine from bench to clinics: Role of food components. Mech Ageing Dev 2024; 220:111960. [PMID: 38971236 DOI: 10.1016/j.mad.2024.111960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/08/2024]
Abstract
Neurodegenerative diseases have multifactorial pathogenesis, mainly involving neuroinflammatory processes. Finding drugs able to treat these diseases, expecially because for most of these diseases there are no effective drugs, and the current drugs cause undesired side effects, represent a crucial point. Most in vivo and in vitro studies have been concentrated on various aspects related to neurons (e.g. neuroprotection), however, there has not been focus on the prevention of early stages involving glial cell activation and neuroinflammation. Recently, it has been demonstrated that nutritional phytochemicals including polyphenols, the main active constituents of the Mediterranean diet, maintain redox balance and neuroprotection through the activation of hormetic vitagene pathway. Recent lipidomics data from our laboratory indicate mushrooms as strong nutritional neuronutrients with strongly activity against neuroinflammation in Meniere' diseaseas, a model of cochleovestibular neural degeneration, as well as in animal model of traumatic brain injury, or rotenone induced parkinson's disease. Moreover, Hidrox®, an aqueous extract of olive containing hydroxytyrosol, and Boswellia, acting as Nrf2 activators, promote resilience by enhancing the redox potential, and thus, regulate through hormetic mechanisms, cellular stress response mechanisms., Thus, modulation of cellular stress pathways, in particular vitagenes system, may be an innovative approach for therapeutic intervention in neurodegenerative disorders.
Collapse
Affiliation(s)
- Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| | - Naomi Osakabe
- Department of Bioscience and Engineering, Shibaura Institute Technology, Tokyo, Japan.
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina 98166, Italy
| | - Sergio Modafferi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, Messina 98168, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina 98166, Italy
| | | | | | - Ali S Abdelhameed
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Luay Rashan
- Biodiversity Unit, Dhofar University, Salalah, Oman
| | - Uwe Wenzel
- Institut für Ernährungswissenschaft, Justus Liebig Universitat Giessen, Germany
| | | | - Edward J Calabrese
- Department of Environmental Health Sciences, Morrill I, N344, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
7
|
Choe JY, Donkor M, Thorpe RJ, Allen MS, Phillips NR, Jones HP. Influence of Diet on Reproducible Corticosterone Levels in a Mouse Model of Maternal Separation with Early Weaning. Life (Basel) 2024; 14:880. [PMID: 39063633 PMCID: PMC11277828 DOI: 10.3390/life14070880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Maternal separation with early weaning (MSEW) is a popular early life stress (ELS) model in rodents, which emulates childhood neglect through scheduled mother-offspring separation. Although variations of ELS models, including maternal separation and MSEW, have been published for the mouse species, the reported results are inconsistent. Corticosterone is considered the main stress hormone involved in regulating stress responses in rodents-yet generating a robust and reproducible corticosterone response in mouse models of ELS has been elusive. Considering the current lack of standardization for MSEW protocols, these inconsistent results may be attributed to variations in model methodologies. Here, we compared the effects of select early wean diet sources-which are the non-milk diets used to complete early weaning in MSEW pups-on the immediate stress phenotype of C57BL/6J mice at postnatal day 21. Non-aversive handling was an integral component of our modified MSEW model. The evaluation of body weight and serum corticosterone revealed the early wean diet to be a key variable in the resulting stress phenotype. Interestingly, select non-milk diets facilitated a stress phenotype in which low body weight was accompanied by significant corticosterone elevation. Our data indicate that dietary considerations are critical in MSEW-based studies and provide insight into improving the reproducibility of key stress-associated outcomes as a function of this widely used ELS paradigm.
Collapse
Affiliation(s)
- Jamie Y. Choe
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX 76107, USA;
- Department of Microbiology, Immunology & Genetics, University of North Texas Health Science Center, Fort Wort, TX 76107, USA; (M.D.); (M.S.A.); (N.R.P.)
| | - Michael Donkor
- Department of Microbiology, Immunology & Genetics, University of North Texas Health Science Center, Fort Wort, TX 76107, USA; (M.D.); (M.S.A.); (N.R.P.)
| | - Roland J. Thorpe
- Hopkins Center for Health Disparities Solutions, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA;
| | - Michael S. Allen
- Department of Microbiology, Immunology & Genetics, University of North Texas Health Science Center, Fort Wort, TX 76107, USA; (M.D.); (M.S.A.); (N.R.P.)
| | - Nicole R. Phillips
- Department of Microbiology, Immunology & Genetics, University of North Texas Health Science Center, Fort Wort, TX 76107, USA; (M.D.); (M.S.A.); (N.R.P.)
- Institute for Translational Research, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Harlan P. Jones
- Department of Microbiology, Immunology & Genetics, University of North Texas Health Science Center, Fort Wort, TX 76107, USA; (M.D.); (M.S.A.); (N.R.P.)
- Institute for Health Disparities, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| |
Collapse
|
8
|
Hofstra BM, Hoeksema EE, Kas MJH, Verbeek DS. Cross-species analysis uncovers the mitochondrial stress response in the hippocampus as a shared mechanism in mouse early life stress and human depression. Neurobiol Stress 2024; 31:100643. [PMID: 38800537 PMCID: PMC11127276 DOI: 10.1016/j.ynstr.2024.100643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Depression, or major depressive disorder, poses a significant burden for both individuals and society, affecting approximately 10.8% of the general population. This psychiatric disorder leads to approximately 800,000 deaths per year. A combination of genetic and environmental factors such as early life stress (ELS) increase the risk for development of depression in humans, and a clear role for the hippocampus in the pathophysiology of depression has been shown. Nevertheless, the underlying mechanisms of depression remain poorly understood, resulting in a lack of effective treatments. To better understand the core mechanisms underlying the development of depression, we used a cross-species design to investigate shared hippocampal pathophysiological mechanisms in mouse ELS and human depression. Mice were subjected to ELS by a maternal separation paradigm, followed by RNA sequencing analysis of the adult hippocampal tissue. This identified persistent transcriptional changes linked to mitochondrial stress response pathways, with oxidative phosphorylation and protein folding emerging as the main mechanisms affected by maternal separation. Remarkably, there was a significant overlap between the pathways involved in mitochondrial stress response we observed and publicly available RNAseq data from hippocampal tissue of depressive patients. This cross-species conservation of changes in gene expression of mitochondria-related genes suggests that mitochondrial stress may play a pivotal role in the development of depression. Our findings highlight the potential significance of the hippocampal mitochondrial stress response as a core mechanism underlying the development of depression. Further experimental investigations are required to expand our understanding of these mechanisms.
Collapse
Affiliation(s)
- Bente M. Hofstra
- Department of Genetics, University of Groningen, University Medical Center Groningen, the Netherlands
- Department of Behavioural Neuroscience, Groningen Institute for Evolutionary Life Sciences, University of Groningen, the Netherlands
| | - Emmy E. Hoeksema
- Department of Behavioural Neuroscience, Groningen Institute for Evolutionary Life Sciences, University of Groningen, the Netherlands
| | - Martien JH. Kas
- Department of Behavioural Neuroscience, Groningen Institute for Evolutionary Life Sciences, University of Groningen, the Netherlands
| | - Dineke S. Verbeek
- Department of Genetics, University of Groningen, University Medical Center Groningen, the Netherlands
| |
Collapse
|
9
|
Sanguino-Gómez J, Krugers HJ. Early-life stress impairs acquisition and retrieval of fear memories: sex-effects, corticosterone modulation, and partial prevention by targeting glucocorticoid receptors at adolescent age. Neurobiol Stress 2024; 31:100636. [PMID: 38883213 PMCID: PMC11177066 DOI: 10.1016/j.ynstr.2024.100636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 03/11/2024] [Accepted: 04/20/2024] [Indexed: 06/18/2024] Open
Abstract
The early postnatal period is a sensitive time window that is characterized by several neurodevelopmental processes that define neuronal architecture and function later in life. Here, we examined in young adult mice, using an auditory fear conditioning paradigm, whether stress during the early postnatal period 1) impacts fear acquisition and memory consolidation in male and female mice; 2) alters the fear responsiveness to corticosterone and 3) whether effects of early-life stress (ELS) can be prevented by treating mice with a glucocorticoid (GR) antagonist at adolescence. Male and female mice were exposed to a limited nesting and bedding model of ELS from postnatal day (PND) 2-9 and injected i.p with RU38486 (RU486) at adolescent age (PND 28-30). At two months of age, mice were trained in the fear conditioning (FC) paradigm (with and without post training administration of corticosterone - CORT) and freezing behavior during fear acquisition and contextual and auditory memory retrieval was scored. We observed that ELS impaired fear acquisition specifically in male mice and reduced both contextual and auditory memory retrieval in male and female mice. Acute post-training administration of CORT increased freezing levels during auditory memory retrieval in female mice but reduced freezing levels during the tone presentation in particular in control males. Treatment with RU486 prevented ELS-effects in acquisition in male mice and in females during auditory memory retrieval. In conclusion, this study highlights the long-lasting consequences of early-life stress on fear memory processing and further illustrates 1) the potential of a glucocorticoid antagonist intervention during adolescence to mitigate these effects and 2) the partial modulation of the auditory retrieval upon post training administration of CORT, with all these effects being sex-dependent.
Collapse
Affiliation(s)
| | - Harm J Krugers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
10
|
Islam R, White JD, Arefin TM, Mehta S, Liu X, Polis B, Giuliano L, Ahmed S, Bowers C, Zhang J, Kaffman A. Early adversity causes sex-specific deficits in perforant pathway connectivity and contextual memory in adolescent mice. Biol Sex Differ 2024; 15:39. [PMID: 38715106 PMCID: PMC11075329 DOI: 10.1186/s13293-024-00616-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 04/26/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Early life adversity impairs hippocampal development and function across diverse species. While initial evidence indicated potential variations between males and females, further research is required to validate these observations and better understand the underlying mechanisms contributing to these sex differences. Furthermore, most of the preclinical work in rodents was performed in adult males, with only few studies examining sex differences during adolescence when such differences appear more pronounced. To address these concerns, we investigated the impact of limited bedding (LB), a mouse model of early adversity, on hippocampal development in prepubescent and adolescent male and female mice. METHODS RNA sequencing, confocal microscopy, and electron microscopy were used to evaluate the impact of LB and sex on hippocampal development in prepubescent postnatal day 17 (P17) mice. Additional studies were conducted on adolescent mice aged P29-36, which included contextual fear conditioning, retrograde tracing, and ex vivo diffusion magnetic resonance imaging (dMRI). RESULTS More severe deficits in axonal innervation and myelination were found in the perforant pathway of prepubescent and adolescent LB males compared to LB female littermates. These sex differences were due to a failure of reelin-positive neurons located in the lateral entorhinal cortex (LEC) to innervate the dorsal hippocampus via the perforant pathway in males, but not LB females, and were strongly correlated with deficits in contextual fear conditioning. CONCLUSIONS LB impairs the capacity of reelin-positive cells located in the LEC to project and innervate the dorsal hippocampus in LB males but not female LB littermates. Given the critical role that these projections play in supporting normal hippocampal function, a failure to establish proper connectivity between the LEC and the dorsal hippocampus provides a compelling and novel mechanism to explain the more severe deficits in myelination and contextual freezing found in adolescent LB males.
Collapse
Affiliation(s)
- Rafiad Islam
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Jordon D White
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Tanzil M Arefin
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, 10016, USA
- Department of Biomedical Engineering, Center for Neurotechnology in Mental Health Research (CNMHR), The Pennsylvania State University, University Park, PA, 16802, USA
| | - Sameet Mehta
- Yale Center for Genomic Analysis, P.O. Box 27386, West Haven, CT, 06516-7386, USA
| | - Xinran Liu
- Department of Cell Biology, Yale University School of Medicine, 333 Cedar Street, SHM IE26, New Haven, CT, 06510, USA
- Center for Cellular and Molecular Imaging, Electron Microscopy Core Facility, Yale University School of Medicine, New Haven, CT, USA
| | - Baruh Polis
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Lauryn Giuliano
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Sahabuddin Ahmed
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Christian Bowers
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA
| | - Jiangyang Zhang
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY, 10016, USA
| | - Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT, 06511, USA.
| |
Collapse
|
11
|
Reisi-Vanani V, Lorigooini Z, Bijad E, Amini-Khoei H. Maternal separation stress through triggering of the neuro-immune response in the hippocampus induces autistic-like behaviors in male mice. Int J Dev Neurosci 2024; 84:87-98. [PMID: 38110192 DOI: 10.1002/jdn.10310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/28/2023] [Accepted: 12/01/2023] [Indexed: 12/20/2023] Open
Abstract
Autism spectrum disorder (ASD) is the fastest-growing neurodevelopmental disease throughout the world. Neuro-immune responses from prenatal to adulthood stages of life induce developmental defects in synaptic signaling, neurotransmitter imbalance, and even structural changes in the brain. In this study, we aimed to focus on the possible role of neuroinflammatory response in the hippocampus in development of the autistic-like behaviors following maternal separation (MS) stress in mice. To do this, mice neonates daily separated from their mothers from postnatal day (PND) 2 to PND 14 for 3 h. During PND45-60, behavioral tests related to autistic-like behaviors including three-chamber sociability, Morris water maze (MWM), shuttle box, resident-intruder, and marble burying tests were performed. Then, hippocampi were dissected out, and the gene expression of inflammatory mediators including TNF-α, IL-1β, TLR4, HMGB1, and NLRP3 was assessed in the hippocampus using RT-PCR. Results showed that MS mice exerted impaired sociability preference, repetitive behaviors, impaired passive avoidance, and spatial memories. The gene expression of inflammatory mediators significantly increased in the hippocampi of MS mice. We concluded that MS stress probably via activating of the HMGB1/TLR4 signaling cascade in the hippocampus induced autistic-like behaviors in mice.
Collapse
Affiliation(s)
- Vahid Reisi-Vanani
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Lorigooini
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Elham Bijad
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hossein Amini-Khoei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
12
|
Ahmed S, Polis B, Jamwal S, Sanganahalli BG, Kaswan ZM, Islam R, Kim D, Bowers C, Giuliano L, Biederer T, Hyder F, Kaffman A. Transient Impairment in Microglial Function Causes Sex-Specific Deficits in Synaptic and Hippocampal Function in Mice Exposed to Early Adversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.14.580284. [PMID: 38405887 PMCID: PMC10888912 DOI: 10.1101/2024.02.14.580284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Abnormal development and function of the hippocampus are two of the most consistent findings in humans and rodents exposed to early life adversity, with males often being more affected than females. Using the limited bedding (LB) paradigm as a rodent model of early life adversity, we found that male adolescent mice that had been exposed to LB exhibit significant deficits in contextual fear conditioning and synaptic connectivity in the hippocampus, which are not observed in females. This is linked to altered developmental refinement of connectivity, with LB severely impairing microglial-mediated synaptic pruning in the hippocampus of male and female pups on postnatal day 17 (P17), but not in adolescent P33 mice when levels of synaptic engulfment by microglia are substantially lower. Since the hippocampus undergoes intense synaptic pruning during the second and third weeks of life, we investigated whether microglia are required for the synaptic and behavioral aberrations observed in adolescent LB mice. Indeed, transient ablation of microglia from P13-21, in normally developing mice caused sex-specific behavioral and synaptic abnormalities similar to those observed in adolescent LB mice. Furthermore, chemogenetic activation of microglia during the same period reversed the microglial-mediated phagocytic deficits at P17 and restored normal contextual fear conditioning and synaptic connectivity in adolescent LB male mice. Our data support an additional contribution of astrocytes in the sex-specific effects of LB, with increased expression of the membrane receptor MEGF10 and enhanced synaptic engulfment in hippocampal astrocytes of 17-day-old LB females, but not in LB male littermates. This finding suggests a potential compensatory mechanism that may explain the relative resilience of LB females. Collectively, these studies highlight a novel role for glial cells in mediating sex-specific hippocampal deficits in a mouse model of early-life adversity.
Collapse
Affiliation(s)
- Sahabuddin Ahmed
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Baruh Polis
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Sumit Jamwal
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Basavaraju G. Sanganahalli
- Department of Radiology & Biomedical Imaging and Magnetic Resonance Research Center, Yale University, New Haven, CT, 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06519, USA
| | - Zoe MacDowell Kaswan
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Rafiad Islam
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Dana Kim
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Christian Bowers
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Lauryn Giuliano
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Thomas Biederer
- Department of Neurology, Yale School of Medicine, 100 College Street, New Haven, CT 06510, USA
| | - Fahmeed Hyder
- Department of Radiology & Biomedical Imaging and Magnetic Resonance Research Center, Yale University, New Haven, CT, 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06519, USA
| | - Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| |
Collapse
|
13
|
Davidson TL, Stevenson RJ. Vulnerability of the Hippocampus to Insults: Links to Blood-Brain Barrier Dysfunction. Int J Mol Sci 2024; 25:1991. [PMID: 38396670 PMCID: PMC10888241 DOI: 10.3390/ijms25041991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
The hippocampus is a critical brain substrate for learning and memory; events that harm the hippocampus can seriously impair mental and behavioral functioning. Hippocampal pathophysiologies have been identified as potential causes and effects of a remarkably diverse array of medical diseases, psychological disorders, and environmental sources of damage. It may be that the hippocampus is more vulnerable than other brain areas to insults that are related to these conditions. One purpose of this review is to assess the vulnerability of the hippocampus to the most prevalent types of insults in multiple biomedical domains (i.e., neuroactive pathogens, neurotoxins, neurological conditions, trauma, aging, neurodegenerative disease, acquired brain injury, mental health conditions, endocrine disorders, developmental disabilities, nutrition) and to evaluate whether these insults affect the hippocampus first and more prominently compared to other brain loci. A second purpose is to consider the role of hippocampal blood-brain barrier (BBB) breakdown in either causing or worsening the harmful effects of each insult. Recent research suggests that the hippocampal BBB is more fragile compared to other brain areas and may also be more prone to the disruption of the transport mechanisms that act to maintain the internal milieu. Moreover, a compromised BBB could be a factor that is common to many different types of insults. Our analysis indicates that the hippocampus is more vulnerable to insults compared to other parts of the brain, and that developing interventions that protect the hippocampal BBB may help to prevent or ameliorate the harmful effects of many insults on memory and cognition.
Collapse
Affiliation(s)
- Terry L. Davidson
- Department of Neuroscience, Center for Neuroscience and Behavior, American University, 4400 Massachusetts Avenue, NW, Washington, DC 20016, USA
| | | |
Collapse
|
14
|
Bao L, Rao J, Yu D, Zheng B, Yin B. Decoding the language of fear: Unveiling objective and subjective indicators in rodent models through a systematic review and meta-analysis. Neurosci Biobehav Rev 2024; 157:105537. [PMID: 38215801 DOI: 10.1016/j.neubiorev.2024.105537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/23/2023] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
While rodent models are vital for studying mental disorders, the underestimation of construct validity of fear indicators has led to limitations in translating to effective clinical treatments. Addressing this gap, we systematically reviewed 5054 articles from the 1960 s, understanding underlying theoretical advancement, and selected 68 articles with at least two fear indicators for a three-level meta-analysis. We hypothesized correlations between different indicators would elucidate similar functions, while magnitude differences could reveal distinct neural or behavioral mechanisms. Our findings reveal a shift towards using freezing behavior as the primary fear indicator in rodent models, and strong, moderate, and weak correlations between freezing and conditioned suppression ratios, 22-kHz ultrasonic vocalizations, and autonomic nervous system responses, respectively. Using freezing as a reference, moderator analysis shows treatment types and fear stages significantly influenced differences in magnitudes between two indicators. Our analysis supports a two-system model of fear in rodents, where objective and subjective fears could operate on a threshold-based mechanism.
Collapse
Affiliation(s)
- Lili Bao
- School of Psychology, Fujian Normal University, China; Key Laboratory for Learning and Behavioral Sciences, Fujian Normal University, China
| | - Jiaojiao Rao
- School of Psychology, Fujian Normal University, China; Key Laboratory for Learning and Behavioral Sciences, Fujian Normal University, China
| | - Delin Yu
- School of Psychology, Fujian Normal University, China; Key Laboratory for Learning and Behavioral Sciences, Fujian Normal University, China
| | - Benhuiyuan Zheng
- School of Psychology, Fujian Normal University, China; Key Laboratory for Learning and Behavioral Sciences, Fujian Normal University, China
| | - Bin Yin
- School of Psychology, Fujian Normal University, China; Key Laboratory for Learning and Behavioral Sciences, Fujian Normal University, China.
| |
Collapse
|
15
|
Salinas-García AF, Roque A, Zamudio-Flores J, Meléndez-Herrera E, Kline AE, Lajud N. Early Life Stress Negatively Impacts Spatial Learning Acquisition and Increases Hippocampal CA1 Microglial Activation After a Mild Traumatic Brain Injury in Adult Male Rats. J Neurotrauma 2024; 41:514-528. [PMID: 37885223 DOI: 10.1089/neu.2023.0452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023] Open
Abstract
Early life stress (ELS) affects neurogenesis and spatial learning, and increases neuroinflammation after a pediatric mild traumatic brain injury (mTBI). Previous studies have shown that ELS has minimal effects in juveniles but shows age-dependent effects in adults. Hence, we aimed to evaluate the effects of ELS in adult male rats after an mTBI. Maternal separation for 180 min per day (MS180) during the first 21 post-natal (P) days was used as the ELS model. At P110, the rats were subjected to a mild controlled cortical impact injury (2.6 mm) or sham surgery. Spatial learning was evaluated in the Morris water maze (MWM) 14 days after surgery and both microglial activation and neurogenesis were quantified. The results indicate that MS180 + mTBI, but not control (CONT) + mTBI, rats show deficiencies in the acquisition of spatial learning. mTBI led to comparable increases in microglial activation in both the hilus and cortical regions for both groups. However, MS180 + mTBI rats exhibited a greater increase in microglial activation in the ipsilateral CA1 hippocampus subfield compared with CONT + mTBI. Interestingly, for the contralateral CA1 region, this effect was observed exclusively in MS180 + mTBI. ELS and mTBI independently caused a decrease in hippocampal neurogenesis and this effect was not increased further in MS180 + mTBI rats. The findings demonstrate that ELS and mTBI synergistically affect cognitive performance and neuroinflammation, thus supporting the hypothesis that increased inflammation resulting from the combination of ELS and mTBI could underlie the observed effects on learning.
Collapse
Affiliation(s)
- Ana Fernanda Salinas-García
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, México
- Instituto de Investigaciones sobre los Recursos Naturales, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, México
| | - Angélica Roque
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, México
- Instituto de Investigaciones sobre los Recursos Naturales, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, México
| | - Jonathan Zamudio-Flores
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, México
- Instituto de Investigaciones sobre los Recursos Naturales, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, México
| | - Esperanza Meléndez-Herrera
- Instituto de Investigaciones sobre los Recursos Naturales, Universidad Michoacana de San Nicolás de Hidalgo, Morelia, Michoacán, México
| | - Anthony E Kline
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, Pennsylvania. USA
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania. USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, Pennsylvania. USA
- Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, Pennsylvania. USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania. USA
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania. USA
| | - Naima Lajud
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Michoacán, México
| |
Collapse
|
16
|
Rodrigues-Ribeiro L, Resende BL, Pinto Dias ML, Lopes MR, de Barros LLM, Moraes MA, Verano-Braga T, Souza BR. Neuroproteomics: Unveiling the Molecular Insights of Psychiatric Disorders with a Focus on Anxiety Disorder and Depression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1443:103-128. [PMID: 38409418 DOI: 10.1007/978-3-031-50624-6_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Anxiety and depression are two of the most common mental disorders worldwide, with a lifetime prevalence of up to 30%. These disorders are complex and have a variety of overlapping factors, including genetic, environmental, and behavioral factors. Current pharmacological treatments for anxiety and depression are not perfect. Many patients do not respond to treatment, and those who do often experience side effects. Animal models are crucial for understanding the complex pathophysiology of both disorders. These models have been used to identify potential targets for new treatments, and they have also been used to study the effects of environmental factors on these disorders. Recent proteomic methods and technologies are providing new insights into the molecular mechanisms of anxiety disorder and depression. These methods have been used to identify proteins that are altered in these disorders, and they have also been used to study the effects of pharmacological treatments on protein expression. Together, behavioral and proteomic research will help elucidate the factors involved in anxiety disorder and depression. This knowledge will improve preventive strategies and lead to the development of novel treatments.
Collapse
Affiliation(s)
- Lucas Rodrigues-Ribeiro
- Department of Physiology and Biophysics, National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Federal University of Minas Gerais, Belo Horizonte, Brazil
- Department of Physiology and Biophysics, Proteomics Group (NPF), Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Bruna Lopes Resende
- Department of Physiology and Biophysics, Laboratory of Neurodevelopment and Evolution (NeuroDEv), Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Maria Luiza Pinto Dias
- Department of Physiology and Biophysics, National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Federal University of Minas Gerais, Belo Horizonte, Brazil
- Department of Physiology and Biophysics, Proteomics Group (NPF), Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Megan Rodrigues Lopes
- Department of Physiology and Biophysics, National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Federal University of Minas Gerais, Belo Horizonte, Brazil
- Department of Physiology and Biophysics, Proteomics Group (NPF), Federal University of Minas Gerais, Belo Horizonte, Brazil
- Department of Physiology and Biophysics, Laboratory of Neurodevelopment and Evolution (NeuroDEv), Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Larissa Luppi Monteiro de Barros
- Department of Physiology and Biophysics, National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Federal University of Minas Gerais, Belo Horizonte, Brazil
- Department of Physiology and Biophysics, Proteomics Group (NPF), Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Muiara Aparecida Moraes
- Department of Physiology and Biophysics, Laboratory of Neurodevelopment and Evolution (NeuroDEv), Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Thiago Verano-Braga
- Department of Physiology and Biophysics, National Institute of Science and Technology in Nanobiopharmaceutics (INCT-Nanobiofar), Federal University of Minas Gerais, Belo Horizonte, Brazil.
- Department of Physiology and Biophysics, Proteomics Group (NPF), Federal University of Minas Gerais, Belo Horizonte, Brazil.
| | - Bruno Rezende Souza
- Department of Physiology and Biophysics, Laboratory of Neurodevelopment and Evolution (NeuroDEv), Federal University of Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
17
|
Brosens N, Simon C, Kessels HW, Lucassen PJ, Krugers HJ. Early life stress lastingly alters the function and AMPA-receptor composition of glutamatergic synapses in the hippocampus of male mice. J Neuroendocrinol 2023; 35:e13346. [PMID: 37901923 DOI: 10.1111/jne.13346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/15/2023] [Accepted: 08/28/2023] [Indexed: 10/31/2023]
Abstract
Early postnatal life is a sensitive period of development that shapes brain structure and function later in life. Exposure to stress during this critical time window can alter brain development and may enhance the susceptibility to psychopathology and neurodegenerative disorders later in life. The developmental effects of early life stress (ELS) on synaptic function are not fully understood, but could provide mechanistic insights into how ELS modifies later brain function and disease risk. We here assessed the effects of ELS on synaptic function and composition in the hippocampus of male mice. Mice were subjected to ELS by housing dams and pups with limited bedding and nesting material from postnatal days (P) 2-9. Synaptic strength was measured in terms of miniature excitatory postsynaptic currents (mEPSCs) in the hippocampal dentate gyrus at three different developmental stages: the early postnatal phase (P9), preadolescence (P21, at weaning) and adulthood at 3 months of age (3MO). Hippocampal synaptosome fractions were isolated from P9 and 3MO tissue and analyzed for protein content to assess postsynaptic composition. Finally, dendritic spine density was assessed in the DG at 3MO. At P9, ELS increased mEPSC frequency and amplitude. In parallel, synaptic composition was altered as PSD-95, GluA3 and GluN2B content were significantly decreased. The increased mEPSC frequency was sustained up to 3MO, at which age, GluA3 content was significantly increased. No differences were found in dendritic spine density. These findings highlight how ELS affects the development of hippocampal synapses, which could provide valuable insight into mechanisms how ELS alters brain function later in life.
Collapse
Affiliation(s)
- Niek Brosens
- SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Carla Simon
- SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Paul J Lucassen
- SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| | - Harm J Krugers
- SILS-CNS, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
18
|
Lonstein JS, Vitale EM, Olekanma D, McLocklin A, Pence N, Bredewold R, Veenema AH, Johnson AW, Burt SA. Anxiety, aggression, reward sensitivity, and forebrain dopamine receptor expression in a laboratory rat model of early-life disadvantage. Dev Psychobiol 2023; 65:e22421. [PMID: 37860907 DOI: 10.1002/dev.22421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/24/2023] [Accepted: 08/17/2023] [Indexed: 10/21/2023]
Abstract
Despite early-life disadvantage (ELD) in humans being a highly heterogenous construct, it consistently predicts negative neurobehavioral outcomes. The numerous environmental contributors and neural mechanisms underlying ELD remain unclear, though. We used a laboratory rat model to evaluate the effects of limited resources and/or heavy metal exposure on mothers and their adult male and female offspring. Dams and litters were chronically exposed to restricted (1-cm deep) or ample (4-cm deep) home cage bedding postpartum, with or without lead acetate (0.1%) in their drinking water from insemination through 1-week postweaning. Restricted-bedding mothers showed more pup-directed behaviors and behavioral fragmentation, while lead-exposed mothers showed more nestbuilding. Restricted bedding-raised male offspring showed higher anxiety and aggression. Either restricted bedding or lead exposure impaired goal-directed performance in a reinforcer devaluation task in females, whereas restricted bedding alone disrupted it in males. Lead exposure, but not limited bedding, also reduced sucrose reward sensitivity in a progressive ratio task in females. D1 and D2 receptor mRNA in the medial prefrontal cortex and nucleus accumbens (NAc) were each affected by the early-life treatments and differently between the sexes. Most notably, adult males (but not females) exposed to both early-life treatments had greatly increased D1 receptor mRNA in the NAc core. These results illuminate neural mechanisms through which ELD threatens neurobehavioral development and highlight forebrain dopamine as a factor.
Collapse
Affiliation(s)
- Joseph S Lonstein
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| | - Erika M Vitale
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| | - Doris Olekanma
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| | - Andrew McLocklin
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| | - Nathan Pence
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| | - Remco Bredewold
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| | - Alexa H Veenema
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| | - Alexander W Johnson
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| | - S Alexandra Burt
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
19
|
Rubinstein MR, Burgueño AL, Quiroga S, Wald MR, Genaro AM. Current Understanding of the Roles of Gut-Brain Axis in the Cognitive Deficits Caused by Perinatal Stress Exposure. Cells 2023; 12:1735. [PMID: 37443769 PMCID: PMC10340286 DOI: 10.3390/cells12131735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/15/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
The term 'perinatal environment' refers to the period surrounding birth, which plays a crucial role in brain development. It has been suggested that dynamic communication between the neuro-immune system and gut microbiota is essential in maintaining adequate brain function. This interaction depends on the mother's status during pregnancy and/or the newborn environment. Here, we show experimental and clinical evidence that indicates that the perinatal period is a critical window in which stress-induced immune activation and altered microbiota compositions produce lasting behavioral consequences, although a clear causative relationship has not yet been established. In addition, we discuss potential early treatments for preventing the deleterious effect of perinatal stress exposure. In this sense, early environmental enrichment exposure (including exercise) and melatonin use in the perinatal period could be valuable in improving the negative consequences of early adversities. The evidence presented in this review encourages the realization of studies investigating the beneficial role of melatonin administration and environmental enrichment exposure in mitigating cognitive alteration in offspring under perinatal stress exposure. On the other hand, direct evidence of microbiota restoration as the main mechanism behind the beneficial effects of this treatment has not been fully demonstrated and should be explored in future studies.
Collapse
Affiliation(s)
- Mara Roxana Rubinstein
- Laboratorio de Psiconeuroendocrinoinmunologia, Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)—Pontificia Universidad Católica Argentina, Buenos Aires C1107AFF, Argentina; (A.L.B.); (S.Q.); (M.R.W.)
| | | | | | | | - Ana María Genaro
- Laboratorio de Psiconeuroendocrinoinmunologia, Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)—Pontificia Universidad Católica Argentina, Buenos Aires C1107AFF, Argentina; (A.L.B.); (S.Q.); (M.R.W.)
| |
Collapse
|
20
|
Hashimoto JG, Singer ML, Goeke CM, Zhang F, Song Y, Xia K, Linhardt RJ, Guizzetti M. Sex differences in hippocampal structural plasticity and glycosaminoglycan disaccharide levels after neonatal handling. Exp Neurol 2023; 361:114313. [PMID: 36572372 PMCID: PMC10097408 DOI: 10.1016/j.expneurol.2022.114313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/05/2022] [Accepted: 12/22/2022] [Indexed: 12/25/2022]
Abstract
In this study we investigated the effects of a neonatal handling protocol that mimics the handling of sham control pups in protocols of neonatal exposure to brain insults on dendritic arborization and glycosaminoglycan (GAG) levels in the developing brain. GAGs are long, unbranched polysaccharides, consisting of repeating disaccharide units that can be modified by sulfation at specific sites and are involved in modulating neuronal plasticity during brain development. In this study, male and female Sprague-Dawley rats underwent neonatal handling daily between post-natal day (PD)4 and PD9, with brains analyzed on PD9. Neuronal morphology and morphometric analysis of the apical and basal dendritic trees of CA1 hippocampal pyramidal neurons were carried out by Golgi-Cox staining followed by neuron tracing and analysis with the software Neurolucida. Chondroitin sulfate (CS)-, Hyaluronic Acid (HA)-, and Heparan Sulfate (HS)-GAG disaccharide levels were quantified in the hippocampus by Liquid Chromatography/Mass Spectrometry analyses. We found sex by neonatal handling interactions on several parameters of CA1 pyramidal neuron morphology and in the levels of HS-GAGs, with females, but not males, showing an increase in both dendritic arborization and HS-GAG levels. We also observed increased expression of glucocorticoid receptor gene Nr3c1 in the hippocampus of both males and females following neonatal handling suggesting that both sexes experienced a similar stress during the handling procedure. This is the first study to show sex differences in two parameters of brain plasticity, CA1 neuron morphology and HS-GAG levels, following handling stress in neonatal rats.
Collapse
Affiliation(s)
- Joel G Hashimoto
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA; VA Portland Health Care System, Portland, OR, USA
| | - Mo L Singer
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA; VA Portland Health Care System, Portland, OR, USA
| | - Calla M Goeke
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA; VA Portland Health Care System, Portland, OR, USA
| | - Fuming Zhang
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Yuefan Song
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Ke Xia
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Robert J Linhardt
- Chemistry and Chemical Biology, Biomedical Engineering, Chemical and Biological Engineering, and Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Marina Guizzetti
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, USA; VA Portland Health Care System, Portland, OR, USA.
| |
Collapse
|
21
|
Dayananda KK, Ahmed S, Wang D, Polis B, Islam R, Kaffman A. Early life stress impairs synaptic pruning in the developing hippocampus. Brain Behav Immun 2023; 107:16-31. [PMID: 36174883 PMCID: PMC10497209 DOI: 10.1016/j.bbi.2022.09.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 09/18/2022] [Accepted: 09/22/2022] [Indexed: 02/08/2023] Open
Abstract
Early life adversity impairs normal hippocampal function and connectivity in various mammalian species, including humans and rodents. According to the 'cumulative model' the number of early adversities can be summed up to determine the risk for developing psychopathology later in life. In contrast, the 'dimensional model' argues that 'Deprivation' and 'Threat' impact different developmental processes that should not be added in determining clinical outcomes. Here we examine these predictions in male and female mice exposed to a single adversity - limited bedding (LB) - versus mice exposed to multiple adversities - unpredictable postnatal stress (UPS) - focusing on microglia-mediated synaptic pruning in the developing hippocampus. Exposure to both LB and UPS reduced the ramification of microglia, impaired their ability to phagocytose synaptic material in vivo and ex vivo, and decreased expression of TREM2. Abnormal phagocytic activity was associated with increased spine density in CA1 pyramidal neurons that was seen in 17-day-old groups and persisted in peri-pubescent 29-day-old LB and UPS mice. Exposure to LB caused more severe impairment in microglial ramification and synaptic engulfment compared to UPS, outcomes that were accompanied by a UPS-specific increase in the expression of several genes implicated in synaptic pruning. We propose that despite being a single stressor, LB represents a more severe form of early deprivation, and that appropriate levels of hippocampal stimulation during the second and third weeks of life are necessary to support normal microglial ramification and synaptic pruning. Further, impaired synaptic pruning during this critical period of hippocampal development contributes to the abnormal hippocampal function and connectivity seen in UPS and LB later in life.
Collapse
Affiliation(s)
- Kiran K Dayananda
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA
| | - Sahabuddin Ahmed
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA
| | - Daniel Wang
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA
| | - Baruh Polis
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA
| | - Rafiad Islam
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA
| | - Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven, CT 06511, USA.
| |
Collapse
|
22
|
Zhu R, Lei Y, Shi F, Tian Q, Zhou X. Arginine Reduces Glycation in γ 2 Subunit of AMPK and Pathologies in Alzheimer's Disease Model Mice. Cells 2022; 11:3520. [PMID: 36359916 PMCID: PMC9655994 DOI: 10.3390/cells11213520] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 09/14/2023] Open
Abstract
UNLABELLED The metabolism disorders are a common convergence of Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM). The characteristics of AD are senile plaques and neurofibrillary tangles (NFTs) composed by deposits of amyloid-β (Aβ) and phosphorylated tau, respectively. Advanced glycation end-products (AGEs) are a stable modification of proteins by non-enzymatic reactions, which could result in the protein dysfunction. AGEs are associated with some disease developments, such as diabetes mellitus and AD, but the effects of the glycated γ2 subunit of AMPK on its activity and the roles in AD onset are unknown. METHODS We studied the effect of glycated γ2 subunit of AMPK on its activity in N2a cells. In 3 × Tg mice, we administrated L-arginine once every two days for 45 days and evaluated the glycation level of γ2 subunit and function of AMPK and alternation of pathologies. RESULTS The glycation level of γ2 subunit was significantly elevated in 3 × Tg mice as compared with control mice, meanwhile, the level of pT172-AMPK was obviously lower in 3 × Tg mice than that in control mice. Moreover, we found that arginine protects the γ2 subunit of AMPK from glycation, preserves AMPK function, and improves pathologies and cognitive deficits in 3 × Tg mice. CONCLUSIONS Arginine treatment decreases glycated γ2 subunit of AMPK and increases p-AMPK levels in 3 × Tg mice, suggesting that reduced glycation of the γ2 subunit could ameliorate AMPK function and become a new target for AD therapy in the future.
Collapse
Affiliation(s)
| | | | | | - Qing Tian
- Key Laboratory of Neurological Disease of Education Ministry, Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xinwen Zhou
- Key Laboratory of Neurological Disease of Education Ministry, Department of Pathophysiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
23
|
Sex-specific effects of neonatal paternal deprivation on microglial cell density in adult California mouse (Peromyscus californicus) dentate gyrus. Brain Behav Immun 2022; 106:1-10. [PMID: 35908654 DOI: 10.1016/j.bbi.2022.07.161] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 11/20/2022] Open
Abstract
Adverse early-life experiences are risk factors for psychiatric disease development, resulting in stress-related neuronal modeling and neurobehavioral changes. Stressful experiences modulate the immune system, contributing to neuronal damage in higher cortical regions, like the hippocampus. Moreover, early-life stressors dysregulate the function of microglia, the resident immune cells of the brain, in the developing hippocampus. Paternal deprivation, an early-life stressor in many biparental species, facilitates sex-dependent inhibitions in hippocampal plasticity, but parental contributors to these sex-specific outcomes are unknown. Also, neurobiological mechanisms contributing to impairments in hippocampal neuroplasticity are less known. Thus, our goals were to 1) determine whether parental behavior is altered in maternal females following removal of the paternal male, 2) assess the effects of paternal deprivation on dentate gyrus (DG) volume and microglia proliferation, and 3) determine if early-life experimental handling mitigates sex-specific reductions in DG cell survival. California mice were born to multiparous breeders and reared by both parents (biparental care) or by their mother alone (i.e., father removed on postnatal day 1; paternal deprivation). One cohort of offspring underwent offspring retrieval tests for eight days beginning on postnatal day 2. On PND 68, these offspring (and a second cohort of mice without behavioral testing) were euthanized and brains visualized for bromodeoxyuridine (BrdU) and neuron-specific class III beta-tubulin (TuJ-1) or ionized calcium binding adaptor molecule 1 (Iba1). While mate absence did not impair maternal retrieval, paternal deprivation reduced DG volume, but Iba1+ cell density was only higher in paternally-deprived females. Neither sex or paternal deprivation significantly altered the number of BrdU+ or Tuj1+ cells in the DG - an absence of a reduction in cell survival may be related to daily handing during early offspring retrieval tests. Together, these data suggest that paternal deprivation impairs hippocampal plasticity; however, sex and early environment may influence the magnitude of these outcomes.
Collapse
|
24
|
Botdorf M, Dunstan J, Sorcher L, Dougherty LR, Riggins T. Socioeconomic disadvantage and episodic memory ability in the ABCD sample: Contributions of hippocampal subregion and subfield volumes. Dev Cogn Neurosci 2022; 57:101138. [PMID: 35907312 PMCID: PMC9335384 DOI: 10.1016/j.dcn.2022.101138] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/22/2022] [Accepted: 07/15/2022] [Indexed: 01/06/2023] Open
Abstract
Socioeconomic disadvantage is associated with volumetric differences in stress-sensitive neural structures, including the hippocampus, and deficits in episodic memory. Rodent studies provide evidence that memory deficits arise via stress-related structural differences in hippocampal subdivisions; however, human studies have only provided limited evidence to support this notion. We used a sample of 10,695 9-13-year-old participants from two timepoints of the Adolescent Brain and Cognitive Development (ABCD) Study to assess whether socioeconomic disadvantage relates to episodic memory performance through hippocampal volumes. We explored associations among socioeconomic disadvantage, measured via the Area Deprivation Index (ADI), concurrent subregion (anterior, posterior) and subfield volumes (CA1, CA3, CA4/DG, subiculum), and episodic memory, assessed via the NIH Toolbox Picture Sequence Memory Test at baseline and 2-year follow-up (Time 2). Results showed that higher baseline ADI related to smaller concurrent anterior, CA1, CA4/DG, and subiculum volumes and poorer Time 2 memory performance controlling for baseline memory. Moreover, anterior, CA1, and subiculum volumes mediated the longitudinal association between the ADI and memory. Results suggest that greater socioeconomic disadvantage relates to smaller hippocampal subregion and subfield volumes and less age-related improvement in memory. These findings shed light on the neural mechanisms linking socioeconomic disadvantage and cognitive ability in childhood.
Collapse
Affiliation(s)
- Morgan Botdorf
- University of Maryland, College Park, Department of Psychology, United States; University of Pennsylvania, Department of Psychology, United States.
| | - Jade Dunstan
- University of Maryland, College Park, Department of Psychology, United States
| | - Leah Sorcher
- University of Maryland, College Park, Department of Psychology, United States
| | - Lea R Dougherty
- University of Maryland, College Park, Department of Psychology, United States
| | - Tracy Riggins
- University of Maryland, College Park, Department of Psychology, United States
| |
Collapse
|
25
|
Culig L, Chu X, Bohr VA. Neurogenesis in aging and age-related neurodegenerative diseases. Ageing Res Rev 2022; 78:101636. [PMID: 35490966 PMCID: PMC9168971 DOI: 10.1016/j.arr.2022.101636] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/14/2022] [Accepted: 04/25/2022] [Indexed: 12/11/2022]
Abstract
Adult neurogenesis, the process by which neurons are generated in certain areas of the adult brain, declines in an age-dependent manner and is one potential target for extending cognitive healthspan. Aging is a major risk factor for neurodegenerative diseases and, as lifespans are increasing, these health challenges are becoming more prevalent. An age-associated loss in neural stem cell number and/or activity could cause this decline in brain function, so interventions that reverse aging in stem cells might increase the human cognitive healthspan. In this review, we describe the involvement of adult neurogenesis in neurodegenerative diseases and address the molecular mechanistic aspects of neurogenesis that involve some of the key aggregation-prone proteins in the brain (i.e., tau, Aβ, α-synuclein, …). We summarize the research pertaining to interventions that increase neurogenesis and regulate known targets in aging research, such as mTOR and sirtuins. Lastly, we share our outlook on restoring the levels of neurogenesis to physiological levels in elderly individuals and those with neurodegeneration. We suggest that modulating neurogenesis represents a potential target for interventions that could help in the fight against neurodegeneration and cognitive decline.
Collapse
Affiliation(s)
- Luka Culig
- Section on DNA Repair, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Xixia Chu
- Section on DNA Repair, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Vilhelm A Bohr
- Section on DNA Repair, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
26
|
Chelini G, Pangrazzi L, Bozzi Y. At the Crossroad Between Resiliency and Fragility: A Neurodevelopmental Perspective on Early-Life Experiences. Front Cell Neurosci 2022; 16:863866. [PMID: 35465609 PMCID: PMC9023311 DOI: 10.3389/fncel.2022.863866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/04/2022] [Indexed: 11/13/2022] Open
Abstract
Postnatal development of the brain is characterized by sensitive windows during which, local circuitry are drastically reshaped by life experiences. These critical periods (CPs) occur at different time points for different brain functions, presenting redundant physiological changes in the underlying brain regions. Although circuits malleability during CPs provides a valuable window of opportunity for adaptive fine-tuning to the living environment, this aspect of neurodevelopment also represents a phase of increased vulnerability for the development of a variety of disorders. Consistently, accumulating epidemiological studies point to adverse childhood experience as a major risk factor for many medical conditions, especially stress- and anxiety-related conditions. Thanks to creative approaches to manipulate rodents’ rearing environment, neurobiologist have uncovered a pivotal interaction between CPs and early-life experiences, offering an interesting landscape to improve our understanding of brain disorders. In this short review, we discuss how early-life experience impacts cellular and molecular players involved in CPs of development, translating into long-lasting behavioral consequences in rodents. Bringing together findings from multiple laboratories, we delineate a unifying theory in which systemic factors dynamically target the maturation of brain functions based on adaptive needs, shifting the balance between resilience and vulnerability in response to the quality of the rearing environment.
Collapse
Affiliation(s)
- Gabriele Chelini
- CIMeC-Center for Mind/Brain Sciences, University of Trento, Rovereto, Italy
- *Correspondence: Gabriele Chelini,
| | - Luca Pangrazzi
- CIMeC-Center for Mind/Brain Sciences, University of Trento, Rovereto, Italy
| | - Yuri Bozzi
- CIMeC-Center for Mind/Brain Sciences, University of Trento, Rovereto, Italy
- Consiglio Nazionale delle Ricerche (CNR) Neuroscience Institute, Pisa, Italy
| |
Collapse
|
27
|
Disrupted presynaptic nectin1-based neuronal adhesion in the entorhinal-hippocampal circuit contributes to early-life stress-induced memory deficits. Transl Psychiatry 2022; 12:141. [PMID: 35379771 PMCID: PMC8980071 DOI: 10.1038/s41398-022-01908-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 01/06/2023] Open
Abstract
The cell adhesion molecule nectin3 and its presynaptic partner nectin1 have been linked to early-life stress-related cognitive disorders, but how the nectin1-nectin3 system contributes to stress-induced neuronal, circuit, and cognitive abnormalities remains to be studied. Here we show that in neonatally stressed male mice, temporal order and spatial working memories, which require the medial entorhinal cortex (MEC)-CA1 pathway, as well as the structural integrity of CA1 pyramidal neurons were markedly impaired in adulthood. These cognitive and structural abnormalities in stressed mice were associated with decreased nectin levels in entorhinal and hippocampal subregions, especially reduced nectin1 level in the MEC and nectin3 level in the CA1. Postnatal suppression of nectin1 but not nectin3 level in the MEC impaired spatial memory, whereas conditional inactivation of nectin1 from MEC excitatory neurons reproduced the adverse effects of early-life stress on MEC-dependent memories and neuronal plasticity in CA1. Our data suggest that early-life stress disrupts presynaptic nectin1-mediated interneuronal adhesion in the MEC-CA1 pathway, which may in turn contribute to stress-induced synaptic and cognitive deficits.
Collapse
|
28
|
Xu B, Zhang X, He Y, Liu C, Li L, Liu Q, Huang Y, Chen M, Ren B, Guo Y, Chen Y. The impacts of early-life adversity on striatal and hippocampal memory functions. Neuroscience 2022; 490:11-24. [DOI: 10.1016/j.neuroscience.2022.02.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/02/2022] [Accepted: 02/25/2022] [Indexed: 12/15/2022]
|
29
|
Sex-specific behavioral and structural alterations caused by early-life stress in C57BL/6 and BTBR mice. Behav Brain Res 2021; 414:113489. [PMID: 34303728 DOI: 10.1016/j.bbr.2021.113489] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/06/2021] [Accepted: 07/21/2021] [Indexed: 11/21/2022]
Abstract
Lately, the development of various mental illnesses, such as depression, personality disorders, and autism spectrum disorders, is often associated with traumatic events in childhood. Nonetheless, the mechanism giving rise to this predisposition is still unknown. Because the development of a disease often depends on a combination of a genetic background and environment, we decided to evaluate the effect of early-life stress on BTBR mice, which have behavioral, neuroanatomical, and physiological features of autism spectrum disorders. As early-life stress, we used prolonged separation of pups from their mothers in the first 2 weeks of life (3 h once a day). We assessed effects of the early-life stress on juvenile (postnatal day 23) and adolescent (postnatal days 37-38) male and female mice of strains C57BL/6 (B6) and BTBR. We found that in both strains, the early-life stress did not lead to changes in the level of social behavior, which is an important characteristic of autism-related behavior. Nonetheless, the early-life stress resulted in increased locomotor activity in juvenile BTBR mice. In adolescent mice, the stress early in life caused a low level of anxiety in B6 males and BTBR females and increased exploratory activity in adolescent BTBR males and females. In addition, adolescent B6 male and female mice with a history of the early-life stress tended to have a thinner motor cortex as assessed by magnetic resonance imaging. As compared to B6 mice, BTBR mice showed reduced levels of social behavior and exploratory activity but their level of locomotor activity was higher. BTBR mice had smaller whole-brain, cortical, and dorsal hippocampal volumes; decreased motor cortex thickness; and increased ventral-hippocampus volume as compared to B6 mice, and these parameters correlated with the level of exploratory behavior of BTBR mice. Overall, the effects of early postnatal stress are sex- and strain-dependent.
Collapse
|