1
|
Snapyan M, Desmeules F, Munro J, Bérard M, Saikali S, Gould PV, Richer M, Pourcher E, Langlois M, Dufresne A, Prud'homme M, Cantin L, Parent A, Saghatelyan A, Parent M. Adult Neurogenesis in the Subventricular Zone of Patients with Huntington's and Parkinson's Diseases and following Long-Term Treatment with Deep Brain Stimulation. Ann Neurol 2025; 97:894-906. [PMID: 39829080 PMCID: PMC12010058 DOI: 10.1002/ana.27181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 01/22/2025]
Abstract
OBJECTIVE Parkinson's and Huntington's diseases are characterized by progressive neuronal loss. Previous studies using human postmortem tissues have shown the impact of neurodegenerative disorders on adult neurogenesis. The extent to which adult neural stem cells are activated in the subventricular zone and whether therapeutic treatments such as deep brain stimulation promote adult neurogenesis remains unclear. The goal of the present study is to assess adult neural stem cells activation and neurogenesis in the subventricular zone of patients with Huntington's and Parkinson's diseases who were treated or not by deep brain stimulation. METHODS Postmortem brain samples from Huntington's and Parkinson's disease patients who had received or not long-term deep brain stimulation of the subthalamic nucleus were used. RESULTS Our results indicate a significant increase in the thickness of the subventricular zone and in the density of proliferating cells and activated stem cells in the brain of Huntington's disease subjects and Parkinson's disease patients treated with deep brain stimulation. We also observed an increase in the density of immature neurons in the brain of these patients. INTERPRETATION Overall, our data indicate that long-term deep brain stimulation of the subthalamic nucleus promotes cell proliferation and neurogenesis in the subventricular zone that are reduced in Parkinson's disease. Taken together, our results also provide a detailed characterization of the cellular composition of the adult human subventricular zone and caudate nucleus in normal condition and in Parkinson's and Huntington's diseases and demonstrate the plasticity of these regions in response to neurodegeneration. ANN NEUROL 2025;97:894-906.
Collapse
Affiliation(s)
| | - Francis Desmeules
- CERVO Brain Research CentreQuebec CityQuebecCanada
- Hôpital de l'Enfant‐Jésus, CHU de Québec‐Université LavalQuebec CityQuebecCanada
| | | | | | - Stephan Saikali
- Hôpital de l'Enfant‐Jésus, CHU de Québec‐Université LavalQuebec CityQuebecCanada
| | - Peter V. Gould
- Hôpital de l'Enfant‐Jésus, CHU de Québec‐Université LavalQuebec CityQuebecCanada
| | - Maxime Richer
- Hôpital de l'Enfant‐Jésus, CHU de Québec‐Université LavalQuebec CityQuebecCanada
| | - Emmanuelle Pourcher
- Hôpital de l'Enfant‐Jésus, CHU de Québec‐Université LavalQuebec CityQuebecCanada
| | - Mélanie Langlois
- Hôpital de l'Enfant‐Jésus, CHU de Québec‐Université LavalQuebec CityQuebecCanada
| | - Anne‐Marie Dufresne
- Hôpital de l'Enfant‐Jésus, CHU de Québec‐Université LavalQuebec CityQuebecCanada
| | - Michel Prud'homme
- Hôpital de l'Enfant‐Jésus, CHU de Québec‐Université LavalQuebec CityQuebecCanada
| | - Léo Cantin
- Hôpital de l'Enfant‐Jésus, CHU de Québec‐Université LavalQuebec CityQuebecCanada
| | - André Parent
- CERVO Brain Research CentreQuebec CityQuebecCanada
- Department of Psychiatry and NeuroscienceFaculty of Medicine, Université LavalQuebec CityQuebecCanada
| | - Armen Saghatelyan
- CERVO Brain Research CentreQuebec CityQuebecCanada
- Department of Psychiatry and NeuroscienceFaculty of Medicine, Université LavalQuebec CityQuebecCanada
- Department of Cellular and Molecular MedicineFaculty of Medicine, University of OttawaOttawaOntarioCanada
| | - Martin Parent
- CERVO Brain Research CentreQuebec CityQuebecCanada
- Department of Psychiatry and NeuroscienceFaculty of Medicine, Université LavalQuebec CityQuebecCanada
| |
Collapse
|
2
|
Lee BH, Eid RS, Hodges TE, Barth C, Galea LAM. Leveraging research into sex differences and steroid hormones to improve brain health. Nat Rev Endocrinol 2025; 21:214-229. [PMID: 39587332 DOI: 10.1038/s41574-024-01061-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 11/27/2024]
Abstract
Sex differences, driven in part by steroid hormones, shape the structure and function of the brain throughout the lifespan and manifest across brain health and disease. The influence of steroid hormones on neuroplasticity, particularly in the adult hippocampus, differs between the sexes, which has important implications for disorders and diseases that compromise hippocampus integrity, such as depression and Alzheimer disease. This Review outlines the intricate relationship between steroid hormones and hippocampal neuroplasticity across the adult lifespan and explores how the unique physiology of male and female individuals can affect health and disease. Despite calls to include sex and gender in research, only 5% of neuroscience studies published in 2019 directly investigated the influence of sex. Drawing on insights from depression, Alzheimer disease and relevant hippocampal plasticity, this Review underscores the importance of considering sex and steroid hormones to achieve a comprehensive understanding of disease susceptibility and mechanisms. Such consideration will enable the discovery of personalized treatments, ultimately leading to improved health outcomes for all.
Collapse
Affiliation(s)
- Bonnie H Lee
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Rand S Eid
- Department of Psychology, McGill University, Montreal, Quebec, Canada
| | - Travis E Hodges
- Department of Psychology and Education, Mount Holyoke College, South Hadley, MA, USA
| | - Claudia Barth
- Division for Mental Health and Substance Abuse, Diakonhjemmet Hospital, Oslo, Norway
| | - Liisa A M Galea
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada.
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
3
|
Downing HC, Glover AB, Gebhardt JE, Thompson KL, Saatman KE. Sex-based differences in the long-term fate of hippocampal neurons born after a traumatic brain injury. Front Behav Neurosci 2025; 19:1523969. [PMID: 39974293 PMCID: PMC11836013 DOI: 10.3389/fnbeh.2025.1523969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/15/2025] [Indexed: 02/21/2025] Open
Abstract
Introduction Moderate-to-severe traumatic brain injury (TBI) results in an early loss of immature hippocampal granule cells and the activation of typically quiescent neural stem cells (NSCs) in the dentate gyrus. Activation of NSCs leads to a robust increase in proliferation and generation of neural progenitor cells (NPCs), supporting restoration of the immature neuron population of over a period of 1-2 weeks. However, it is unclear if neurons born early after injury develop normally, survive long-term and functionally integrate into the hippocampal network. Although adult hippocampal neurogenesis is regulated in a sex-dependent manner, the majority of pre-clinical TBI studies lack the inclusion of both sexes. The goal of this study was to examine sex differences in hippocampal neurogenesis in response to a moderate controlled cortical impact brain injury. Methods In-vivo labeling of NPCs and tracking of their morphological development into a granule cell was achieved using an inducible Cre recombinase driven by the Ascl1 promoter in a CAG-floxStopTom reporter mouse. Ascl1 is a basic helix-loop-helix transcription factor transiently expressed in NPCs and activated NSCs in the dentate gyrus of the adult mammalian brain. To specifically label NPCs born acutely after TBI, tamoxifen was delivered to mice on days 2 and 3 postinjury. Mice survived to 6 weeks after TBI to allow for full neuronal maturation of tdTomato-labeled NPCs. Results At 6 weeks postinjury, numbers of tdTomato-positive granule cells were significantly reduced in the ipsilateral hippocampus of brain-injured mice compared to controls, with a more pronounced decrease in males. Further, posttrauma-born neurons in males, but not females, exhibited impaired dendritic development. Neurons born after injury extended axons which formed synaptic terminals within the CA3 region. Numbers of mossy fiber boutons were significantly decreased in injured males compared to naïve males or to injured females. Potential forms of plasticity were observed in brain-injured females, including increased neurogenesis in the contralateral hippocampus and increased mossy fiber bouton volume. Together these data suggest a neurogenic advantage in females after injury. Discussion This study is the first to report sex differences in posttraumatic hippocampal neurogenesis and to demonstrate modification of synaptic terminals formed by neurons born after TBI.
Collapse
Affiliation(s)
- Hannah C. Downing
- Department of Physiology, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
| | - Ashley B. Glover
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
| | - Jessica E. Gebhardt
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Katherine L. Thompson
- Dr. Bing Zhang Department of Statistics, College of Arts and Sciences, University of Kentucky, Lexington, KY, United States
| | - Kathryn E. Saatman
- Department of Physiology, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
4
|
Lee BH, Cevizci M, Lieblich SE, Galea LAM. Sex-specific influences of APOEε4 genotype on hippocampal neurogenesis and progenitor cells in middle-aged rats. Biol Sex Differ 2025; 16:10. [PMID: 39910616 DOI: 10.1186/s13293-025-00694-8] [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: 12/01/2024] [Accepted: 01/27/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) disproportionately and uniquely affects females, and these sex differences are further exacerbated by the presence of Apolipoprotein (APOE) ε4 alleles, the top genetic risk factor for late-onset AD. To expand our understanding about how late-onset AD risk might differentially influence males and females, this study explores how APOEε4 affects hippocampal neurogenesis and microglia, key neuroplastic markers involved in AD pathogenesis, differently by sex in middle-aged rats. METHODS A rat model expressing the humanized (h) APOEε4 allele was characterized to examine markers of adult neurogenesis (neural progenitor cells and new-born neurons) and immune cells (microglia) in the dentate gyrus of the hippocampus in 13 month-old male and female rats. RESULTS We observed basal sex differences in neurogenesis at middle age, as wildtype male rats had greater densities of neural progenitor cells and new-born neurons in the dentate gyrus than wildtype female rats. Male hAPOEε4 rats exhibited fewer neural progenitor cells, fewer new-born neurons, and more microglia than male wildtype rats. On the other hand, female hAPOEε4 rats exhibited more new-born neurons than female wildtype rats. Interestingly, females had more microglia than males regardless of genotype. Correlations were conducted to further elucidate any sex differences in the relationships between these biomarkers. Notably, there was a significant positive correlation between neural progenitor cells and new-born neurons, and a significant negative correlation between new-born neurons and microglia, but only in male rats. CONCLUSION In contrast to the clear pattern of effects of the hAPOEε4 risk factor on hippocampal neurogenesis in males, females had unaltered levels of neural progenitor cells and increased density of new-born neurons. Furthermore, relationships between neurogenesis and microglia were significantly correlated within males, and not females. This suggests that females may be presenting a compensatory response to the hAPOEε4 genotype at middle age. Collectively, these results exemplify the importance of thoroughly examining influences of sex on AD endophenotypes, as it may reveal sex-specific pathways and protective mechanisms relevant to AD.
Collapse
Affiliation(s)
- Bonnie H Lee
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Melike Cevizci
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie E Lieblich
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada.
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada.
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada.
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
5
|
Sullens DG, Gilley K, Moraglia LE, Dison S, Hoffman JT, Wiffler MB, Barnes RC, Ginty AT, Sekeres MJ. Sex in aging matters: exercise and chronic stress differentially impact females and males across the lifespan. Front Aging Neurosci 2025; 16:1508801. [PMID: 39881679 PMCID: PMC11774976 DOI: 10.3389/fnagi.2024.1508801] [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: 10/09/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Assessing sex as a biological variable is critical to determining the influence of environmental and lifestyle risks and protective factors mediating behavior and neuroplasticity across the lifespan. We investigated sex differences in affective behavior, memory, and hippocampal neurogenesis following short- or long-term exposure to exercise or chronic mild stress in young and aged mice. Male and female mice were assigned control, running, or chronic stress rearing conditions for 1 month (young) or for 15 months (aged), then underwent a behavioral test battery to assess activity, affective behavior, and memory. Stress exposure into late-adulthood increased hyperactivity in both sexes, and enhanced anxiety-like and depressive-like behavior in aged female, but not male, mice. One month of stress or running had no differential effects on behavior in young males and females. Running increased survival of BrdU-labelled hippocampal cells in both young and aged mice, and enhanced spatial memory in aged mice. These findings highlight the importance of considering sex when determining how aging is differently impacted by modifiable lifestyle factors across the lifespan.
Collapse
Affiliation(s)
- D. Gregory Sullens
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, United States
| | - Kayla Gilley
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, United States
- Department of Biology and Chemistry, Liberty University, Lynchburg, VA, United States
| | - Luke E. Moraglia
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, United States
- Department of Psychology, The University of Texas at Dallas, Richardson, TX, United States
| | - Sarah Dison
- Department of Biology, Baylor University, Waco, TX, United States
| | - Jessica T. Hoffman
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, United States
| | - Madison B. Wiffler
- Department of Biology, Baylor University, Waco, TX, United States
- Department of Neurobiology, University of Utah, Salt Lake City, UT, United States
| | - Robert C. Barnes
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, United States
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Annie T. Ginty
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, United States
| | - Melanie J. Sekeres
- Department of Psychology and Neuroscience, Baylor University, Waco, TX, United States
- School of Psychology, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
6
|
Weston NM, Green JC, Keoprasert TN, Sun D. Dendritic morphological development of traumatic brain injury-induced new neurons in the dentate gyrus is important for post-injury cognitive recovery and is regulated by Notch1. Exp Neurol 2024; 382:114963. [PMID: 39303845 PMCID: PMC11502241 DOI: 10.1016/j.expneurol.2024.114963] [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: 07/09/2024] [Revised: 08/27/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024]
Abstract
Traumatic brain injury (TBI) is a prevalent problem with survivors suffering from chronic cognitive impairments. Following TBI there is a series of neuropathological changes including neurogenesis. It is well established that neurogenesis in the dentate gyrus (DG) of the hippocampus is important for hippocampal dependent learning and memory functions. Following TBI, injury-enhanced hippocampal neurogenesis is believed to contribute to post-injury cognitive recovery. Behavioral function is connected to synaptic plasticity and neuronal dendritic branching is critical for successful synapse formation. To ascertain the functional contribution of injury-induced DG new neurons in post-TBI cognitive recovery, it is necessary to study their dendritic morphological development and the molecular mechanisms controlling this process. Utilizing transgenic mice with tamoxifen-induced GFP expression and Notch1 knock-out in nestin+ neural stem cells, this study examined dendritic morphology, the role of Notch1 in regulating dendritic complexity of injury-induced DG new neurons, and their association to post-TBI cognitive recovery. We found that at 8 weeks after a moderate TBI, injury-induced DG new neurons in the injured control mice displayed a similar dendritic morphology as the cells in non-injured mice accompanied with cognitive recovery. In comparison, in Notch1 conditional knock-out mice, DG new neurons in the injured mice had a significant reduction in dendritic morphological development including dendritic arbors, volume span, and number of branches in comparison to the cells in non-injured mice concomitant with persistent cognitive dysfunction. The results of this study confirm the importance of post-injury generated new neurons in cognitive recovery following TBI and the role of Notch1 in regulating their maturation process.
Collapse
Affiliation(s)
- Nicole M Weston
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, United States
| | - Jakob C Green
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, United States
| | - Timothy N Keoprasert
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, United States
| | - Dong Sun
- Department of Anatomy and Neurobiology, School of Medicine, Virginia Commonwealth University, United States.
| |
Collapse
|
7
|
Hillerer KM, Gimsa U. Adult neurogenesis and the microbiota-gut-brain axis in farm animals: underestimated and understudied parameters for improving welfare in livestock farming. Front Neurosci 2024; 18:1493605. [PMID: 39664450 PMCID: PMC11631930 DOI: 10.3389/fnins.2024.1493605] [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: 09/09/2024] [Accepted: 11/05/2024] [Indexed: 12/13/2024] Open
Abstract
Welfare in commercial livestock farming is becoming increasingly important in current agriculture research. Unfortunately, there is a lack of understanding about the neuronal mechanisms that underlie well-being on an individual level. Neuroplasticity in the hippocampus, the subventricular zone (SVZ), the olfactory bulb (OB) and the hypothalamus may be essential regulatory components in the context of farm animal behaviour and welfare that may be altered by providing environmental enrichment (EE). The importance of pre-and probiotics as a form of EE and the microbiota-gut-brain axis (MGBA) has come under the spotlight in the last 20 years, particularly in the contexts of research into stress and of stress resilience. However, it could also be an important regulatory system for animal welfare in livestock farming. This review aims to present a brief overview of the effects of EE on physiology and behaviour in farm animals and briefly discusses literature on behavioural flexibility, as well as inter-individual stress-coping styles and their relationship to animal welfare. Most importantly, we will summarise the literature on different forms of neural plasticity in farm animals, focusing on neurogenesis in various relevant brain regions. Furthermore, we will provide a brief outlook connecting these forms of neuroplasticity, stress, EE, the MGBA and welfare measures in modern livestock farming, concentrating on pigs.
Collapse
Affiliation(s)
- Katharina M. Hillerer
- Research Institute for Farm Animal Biology (FBN), Dummerstorf, Mecklenburg-Vorpommern, Germany
| | | |
Collapse
|
8
|
Yagi S, Mohammad A, Wen Y, Batallán Burrowes AA, Blankers SA, Galea LAM. Estrogens dynamically regulate neurogenesis in the dentate gyrus of adult female rats. Hippocampus 2024; 34:583-597. [PMID: 39166359 DOI: 10.1002/hipo.23633] [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: 01/26/2024] [Revised: 08/01/2024] [Accepted: 08/09/2024] [Indexed: 08/22/2024]
Abstract
Estrone and estradiol differentially modulate neuroplasticity and cognition. How they influence the maturation of new neurons in the adult hippocampus, however, is not known. The present study assessed the effects of estrone and estradiol on the maturation timeline of neurogenesis in the dentate gyrus (DG) of ovariectomized (a model of surgical menopause) young adult Sprague-Dawley rats using daily subcutaneous injections of 17β-estradiol, estrone or vehicle. Rats were injected with a DNA synthesis marker, 5-bromo-2-deoxyuridine (BrdU), and were perfused 1, 2, or 3 weeks after BrdU injection and daily hormone treatment. Brains were sectioned and processed for various markers including: sex-determining region Y-box 2 (Sox2), glial fibrillary acidic protein (GFAP), antigen kiel 67 (Ki67), doublecortin (DCX), and neuronal nuclei (NeuN). Immunofluorescent labeling or co-labelling of BrdU with Sox2 (progenitor cells), Sox2/GFAP (neural progenitor cells), Ki67 (cell proliferation), DCX (immature neurons), NeuN (mature neurons) was used to examine the trajectory and maturation of adult-born neurons over time. Estrogens had early (1 week of exposure) effects on different stages of neurogenesis (neural progenitor cells, cell proliferation and early maturation of new cells into neurons) but these effects were less pronounced after prolonged treatment. Estradiol enhanced, whereas estrone reduced cell proliferation after 1 week but not after longer exposure to either estrogen. Both estrogens increased the density of immature neurons (BrdU/DCX-ir) after 1 week of exposure compared to vehicle treatment but this increased density was not sustained over longer durations of treatments to estrogens, suggesting that the enhancing effects of estrogens on neurogenesis were short-lived. Longer duration post-ovariectomy, without treatments with either of the estrogens, was associated with reduced neural progenitor cells in the DG. These results demonstrate that estrogens modulate several aspects of adult hippocampal neurogenesis differently in the short term, but may lose their ability to influence neurogenesis after long-term exposure. These findings have potential implications for treatments involving estrogens after surgical menopause.
Collapse
Affiliation(s)
- Shunya Yagi
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ahmad Mohammad
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Yanhua Wen
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ariel A Batallán Burrowes
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Samantha A Blankers
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Zhou JZ, Deng J, Luo DX, Mai JW, Wu JY, Duan YJ, Dong B, Xin WJ, Xu T, Wei JY. Sex differences in functional and structural alterations of hippocampus region in chronic pain: a DTI and resting-state fMRI study. Front Neurosci 2024; 18:1428666. [PMID: 39308951 PMCID: PMC11412943 DOI: 10.3389/fnins.2024.1428666] [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: 05/06/2024] [Accepted: 08/15/2024] [Indexed: 09/25/2024] Open
Abstract
Introduction It is well known that there are significant differences in the prevalence of chronic pain between males and females. Human and animal imaging studies have shown that chronic pain profoundly alters the structure and function of brain regions. However, there is limited research on the sex-specific mechanisms underlying the brain plasticity and adaptive changes associated with chronic pain. In this article, we conducted a multimodal study to evaluate how nerve injury-induced chronic pain affects the brain. Methods Male and female Sprague-Dawley (SD) rats with spared nerve injury (SNI) model underwent resting-state functional magnetic resonance imaging (rs-fMRI) (male sham group: n = 18; male SNI group: n = 18; female sham group: n = 20; female SNI group: n = 18) and magnetic resonance diffusion tensor imaging (DTI) (male sham group: n = 23; male SNI group: n = 21; female sham group: n = 20; female SNI group: n = 21) scanning. ICA method, Fractional amplitude of low-frequency fluctuations (fALFF), immunofluorescence staining, and graph theory analysis was utilized to extract the rs-fMRI changes of brain regions of each group. Results Using SNI model, which promotes long-lasting mechanical allodynia, we found that neuropathic pain deeply modified the intrinsic organization of the brain functional network in male and female rats (main effect of operation: F = 298.449, P < 0.001). 64 independent components (ICs) in the brain were divided and assigned to 16 systems. In male rats, we observed significant alterations in the microstructure of the hippocampal cornu ammonis 1 and cornu ammonis 2 (CA1/CA2) region, as indicated by increased mean diffusivity (MD) (CA1_L: P = 0.02; CA1_R: P = 0.031; CA2_L: P = 0.035; CA2_R: P = 0.015) and radial diffusivity (RD) (CA1_L: P = 0.028; CA1_R: P = 0.033; CA2_L: P = 0.037; CA2_R: P = 0.038) values, along with enhanced activating transcription factor 3 (ATF3) expression. Conversely, in female rats, we found significant increases in the fractional amplitude of low frequency fluctuations (fALFF) value within the hippocampal dentate gyrus (DG) (F = 5.419, P = 0.023), accompanied by elevated c-Fos signal (F = 6.269, P = 0.031). Furthermore, graph theory analysis revealed notable differences in the small-world network of the hippocampal system in female rats, characterized by reduced small-world attributes and increased inter-nodal transmission efficiency. Discussion Our study indicates sex differences in structural and functional alterations in the hippocampal system in rats under chronic pain conditions. The results suggest that the hippocampus system plays an important role in the different mechanisms of chronic pain in different sexes. These findings provide reliable insights to explore the complex mechanisms underlying sex differences in chronic pain.
Collapse
Affiliation(s)
- Jun-Zhi Zhou
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
- Neuroscience Program, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Department of Physiology and Pain Research Center, Sun Yat-sen University, Guangzhou, China
| | - Jie Deng
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
- Neuroscience Program, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Department of Physiology and Pain Research Center, Sun Yat-sen University, Guangzhou, China
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - De-Xing Luo
- Department of Anesthesiology, Huizhou Central People’s Hospital, Huizhou, China
| | - Jing-Wen Mai
- Department of Anesthesiology, Huizhou Central People’s Hospital, Huizhou, China
| | - Jia-Yan Wu
- Neuroscience Program, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Department of Physiology and Pain Research Center, Sun Yat-sen University, Guangzhou, China
| | - Yu-Juan Duan
- Neuroscience Program, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Department of Physiology and Pain Research Center, Sun Yat-sen University, Guangzhou, China
| | - Bo Dong
- Neuroscience Program, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Department of Physiology and Pain Research Center, Sun Yat-sen University, Guangzhou, China
| | - Wen-Jun Xin
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
- Neuroscience Program, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Department of Physiology and Pain Research Center, Sun Yat-sen University, Guangzhou, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Ting Xu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
- Neuroscience Program, Zhongshan School of Medicine, Guangdong Province Key Laboratory of Brain Function and Disease, Department of Physiology and Pain Research Center, Sun Yat-sen University, Guangzhou, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Jia-You Wei
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Zhuhai, China
| |
Collapse
|
10
|
Lee BH, Cevizci M, Lieblich SE, Ibrahim M, Wen Y, Eid RS, Lamers Y, Duarte-Guterman P, Galea LAM. Exploring the parity paradox: Differential effects on neuroplasticity and inflammation by APOEe4 genotype at middle age. Brain Behav Immun 2024; 120:54-70. [PMID: 38772427 DOI: 10.1016/j.bbi.2024.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 04/20/2024] [Accepted: 05/18/2024] [Indexed: 05/23/2024] Open
Abstract
Female sex and Apolipoprotein E (APOE) ε4 genotype are top non-modifiable risk factors for Alzheimer's disease (AD). Although female-unique experiences like parity (pregnancy and motherhood) have positive effects on neuroplasticity at middle age, previous pregnancy may also contribute to AD risk. To explore these seemingly paradoxical long-term effects of parity, we investigated the impact of parity with APOEε4 genotype by examining behavioural and neural biomarkers of brain health in middle-aged female rats. Our findings show that primiparous (parous one time) hAPOEε4 rats display increased use of a non-spatial cognitive strategy and exhibit decreased number and recruitment of new-born neurons in the ventral dentate gyrus of the hippocampus in response to spatial working memory retrieval. Furthermore, primiparity and hAPOEε4 genotype synergistically modulate inflammatory markers in the ventral hippocampus. Collectively, these findings demonstrate that previous parity in hAPOEε4 rats confers an added risk to present with reduced activity and engagement of the hippocampus as well as elevated pro-inflammatory signaling, and underscore the importance of considering female-specific factors and genotype in health research.
Collapse
Affiliation(s)
- Bonnie H Lee
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Melike Cevizci
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie E Lieblich
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Muna Ibrahim
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Yanhua Wen
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Rand S Eid
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Yvonne Lamers
- Food Nutrition and Health Program, Faculty of Land and Food Systems, University of British Columbia, Vancouver, BC, Canada
| | - Paula Duarte-Guterman
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, ON, Canada.
| |
Collapse
|
11
|
Bradshaw JL, Wilson EN, Mabry S, Shrestha P, Gardner JJ, Cunningham RL. Impact of sex and hypoxia on brain region-specific expression of membrane androgen receptor AR45 in rats. Front Endocrinol (Lausanne) 2024; 15:1420144. [PMID: 39092288 PMCID: PMC11291194 DOI: 10.3389/fendo.2024.1420144] [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: 04/19/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
Background Sex differences in oxidative stress-associated cognitive decline are influenced by sex hormone levels. Notably, oxidative stress-associated neuronal cell death can be exacerbated through testosterone signaling via membrane androgen receptor AR45, which is complexed with G protein Gαq within plasma membrane-associated lipid rafts. The objective of this study was to elucidate the impact of sex on the expression of AR45 and Gαq in brain regions associated with cognitive function, specifically hippocampus subregions and entorhinal cortex. Additionally, we investigated whether chronic intermittent hypoxia (CIH), an oxidative stressor with sex-specific effects, would modulate AR45 and Gαq expression in these brain regions. Methods Adult male and female Sprague-Dawley rats were exposed to CIH or normoxia (room air) during their sleep phase for 14 days. We quantified AR45 and Gαq protein expression in various cognition-associated brain regions [dorsal hippocampal CA1, CA3, dentate gyrus (DG), and entorhinal cortex (ETC)] via western blotting. For comparisons, AR45 and Gαq protein expression were also assessed in brain regions outside the hippocampal-ETC circuit [thalamus (TH) and striatum (STR)]. Results The highest AR45 levels were expressed in the hippocampal CA1 and DG while the lowest expression was observed in the extrahippocampal STR. The highest Gαq levels were expressed in the hippocampal-associated ETC while the lowest expression was observed in the extrahippocampal TH. Females expressed higher levels of AR45 in the hippocampal DG compared to males, while no sex differences in Gαq expression were observed regardless of brain region assessed. Moreover, there was no effect of CIH on AR45 or Gαq expression in any of the brain regions examined. AR45 expression was positively correlated with Gαq expression in the CA1, DG, ETC, TH, and STR in a sex-dependent manner. Conclusion Our findings reveal enrichment of AR45 and Gαq protein expression within the hippocampal-ETC circuit, which is vulnerable to oxidative stress and neurodegeneration during cognitive decline. Nonetheless, CIH does not modulate the expression of AR45 or Gαq. Importantly, there are sex differences in AR45 expression and its association with Gαq expression in various brain regions, which may underlie sex-specific differences in cognitive and motor function-associated declines with aging.
Collapse
Affiliation(s)
- Jessica L. Bradshaw
- Department of Pharmaceutical Sciences, University of North Texas (UNT) System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - E. Nicole Wilson
- Department of Pharmaceutical Sciences, University of North Texas (UNT) System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Steve Mabry
- Department of Pharmaceutical Sciences, University of North Texas (UNT) System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Pawan Shrestha
- Department of Pharmaceutical Sciences, University of North Texas (UNT) System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, United States
- North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Jennifer J. Gardner
- Department of Pharmaceutical Sciences, University of North Texas (UNT) System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Rebecca L. Cunningham
- Department of Pharmaceutical Sciences, University of North Texas (UNT) System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, United States
| |
Collapse
|
12
|
Luo Y, Wang Y, Qiu F, Hou G, Liu J, Yang H, Wu M, Dong X, Guo D, Zhong Z, Zhang X, Ge J, Meng P. Ablated Sonic Hedgehog Signaling in the Dentate Gyrus of the Dorsal and Ventral Hippocampus Impairs Hippocampal-Dependent Memory Tasks and Emotion in a Rat Model of Depression. Mol Neurobiol 2024; 61:4352-4368. [PMID: 38087166 DOI: 10.1007/s12035-023-03796-9] [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: 08/09/2023] [Accepted: 11/08/2023] [Indexed: 07/11/2024]
Abstract
Specific memory processes and emotional aberrations in depression can be attributed to the different dorsal-ventral regions of the hippocampus. However, the molecular mechanisms underlying the differential functions of the dorsal hippocampus (dHip) and ventral hippocampus (vHip) remain unclear. As Sonic Hedgehog (Shh) is involved in the dorsal-ventral patterning of the neural tube and its signaling is dysregulated by chronic unpredictable mild stress (CUMS), we investigated its role in influencing the differential functions of the dHip and vHip. Here, CUMS downregulated the expression of Shh signaling markers, including Shh and its downstream effectors GLI family zinc finger 12 (Gli1/2), Patched (Ptch), and smoothened (Smo), in both the dHip and vHip of rats, though more so in the vHip. Additionally, Shh knockdown in the dorsal or ventral dentate gyrus (DG) resulted in restrained neurogenic activity in newborn neurons, especially in immature neurons through decreased expression of Shh signaling markers. Furthermore, Shh knockdown in the DG of the dHip led to memory impairment by inhibiting experience-dependent activation of immature neurons, whereas its knockdown in the DG of the vHip led to an emotional handicap by delaying the maturation of immature neurons. Finally, Shh knockdown in either the dDG or vDG of hippocampus abolished the corresponding cognitive enhancement and emotional recovery of fluoxetine. In conclusion, Shh is essential to maintain the functional heterogeneity of dHip and vHip in depressed rat, which was mainly mediating by local changes of dependent activation and maturity of immature neurons, respectively.
Collapse
Affiliation(s)
- Yan Luo
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Yan Wang
- Yiyang Central Hospital, Yiyang, 413000, Hunan, China
| | - Feng Qiu
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Guanghan Hou
- Fourth Hospital of Changsha, Hunan, 410000, China
| | - Jian Liu
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
- First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China
| | - Hui Yang
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
- First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, 410007, Hunan, China
| | - Mei Wu
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Xuanqi Dong
- The Second People's Hospital of Hunan Province, Changsha, 410000, Hunan, China
| | - Dongwei Guo
- The Second People's Hospital of Hunan Province, Changsha, 410000, Hunan, China
| | - Ziyan Zhong
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Xi Zhang
- The Second People's Hospital of Hunan Province, Changsha, 410000, Hunan, China.
| | - Jinwen Ge
- Hunan Academy of Chinese Medicine, Changsha, 410300, Hunan, China.
| | - Pan Meng
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
13
|
Beltran-Casanueva R, Hernández-García A, Serrano-Castro PJ, Sánchez-Pérez JA, Barbancho-Fernández MA, García-Casares N, Fuxe K, Borroto-Escuela DO, Narváez M. Long-term enhancements in antidepressant efficacy and neurogenesis: Effects of intranasal co-administration of neuropeptide Y 1 receptor (NPY1R) and galanin receptor 2 (GALR2) agonists in the ventral hippocampus. FASEB J 2024; 38:e23595. [PMID: 38572811 DOI: 10.1096/fj.202400087r] [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: 01/13/2024] [Revised: 02/27/2024] [Accepted: 03/25/2024] [Indexed: 04/05/2024]
Abstract
This study evaluates the sustained antidepressant-like effects and neurogenic potential of a 3-day intranasal co-administration regimen of galanin receptor 2 (GALR2) agonist M1145 and neuropeptide Y Y1 receptor (NPY1R) agonist [Leu31, Pro34]NPY in the ventral hippocampus of adult rats, with outcomes analyzed 3 weeks post-treatment. Utilizing the forced swimming test (FST), we found that this co-administration significantly enhances antidepressant-like behaviors, an effect neutralized by the GALR2 antagonist M871, highlighting the synergistic potential of these neuropeptides in modulating mood-related behaviors. In situ proximity ligation assay (PLA) indicated a significant increase in GALR2/NPYY1R heteroreceptor complexes in the ventral hippocampal dentate gyrus, suggesting a molecular basis for the behavioral outcomes observed. Moreover, proliferating cell nuclear antigen (PCNA) immunolabeling revealed increased cell proliferation in the subgranular zone of the dentate gyrus, specifically in neuroblasts as evidenced by co-labeling with doublecortin (DCX), without affecting quiescent neural progenitors or astrocytes. The study also noted a significant uptick in the number of DCX-positive cells and alterations in dendritic morphology in the ventral hippocampus, indicative of enhanced neuronal differentiation and maturation. These morphological changes highlight the potential of these agonists to facilitate the functional integration of new neurons into existing neural circuits. By demonstrating the long-lasting effects of a brief, 3-day intranasal administration of GALR2 and NPY1R agonists, our findings contribute significantly to the understanding of neuropeptide-mediated neuroplasticity and herald novel therapeutic strategies for the treatment of depression and related mood disorders, emphasizing the therapeutic promise of targeting neurogenesis and neuronal maturation processes.
Collapse
Affiliation(s)
- Rasiel Beltran-Casanueva
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Receptomics and Brain Disorders Lab, Edificio Lopez-Peñalver, Instituto de Investigación Biomédica de Málaga, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Aracelis Hernández-García
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Receptomics and Brain Disorders Lab, Edificio Lopez-Peñalver, Instituto de Investigación Biomédica de Málaga, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
- Departamento de Docencia e Investigación, Universidad de Ciencias Médicas de Holguín, Hospital Pedíatrico Universitario Octavio de la Concepción de la Pedraja, Holguín, Cuba
| | - Pedro Jesús Serrano-Castro
- Instituto de Investigación Biomédica de Málaga, NeuronLab, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga, Unit of Neurology, Hospital Regional Universitario de Málaga, Málaga, Spain
- Vithas Málaga, Grupo Hospitalario Vithas, Málaga, Spain
| | - Jose Andrés Sánchez-Pérez
- Instituto de Investigación Biomédica de Málaga, NeuronLab, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga, Unit of Psychiatry, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | | | - Natalia García-Casares
- Instituto de Investigación Biomédica de Málaga, NeuronLab, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Dasiel O Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Receptomics and Brain Disorders Lab, Edificio Lopez-Peñalver, Instituto de Investigación Biomédica de Málaga, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Manuel Narváez
- Instituto de Investigación Biomédica de Málaga, NeuronLab, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga, Unit of Neurology, Hospital Regional Universitario de Málaga, Málaga, Spain
- Vithas Málaga, Grupo Hospitalario Vithas, Málaga, Spain
| |
Collapse
|
14
|
Borroto-Escuela D, Serrano-Castro P, Sánchez-Pérez JA, Barbancho-Fernández MA, Fuxe K, Narváez M. Enhanced neuronal survival and BDNF elevation via long-term co-activation of galanin 2 (GALR2) and neuropeptide Y1 receptors (NPY1R): potential therapeutic targets for major depressive disorder. Expert Opin Ther Targets 2024; 28:295-308. [PMID: 38622072 DOI: 10.1080/14728222.2024.2342517] [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: 03/01/2024] [Accepted: 04/05/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND Major Depressive Disorder (MDD) is a prevalent and debilitating condition, necessitating novel therapeutic strategies due to the limited efficacy and adverse effects of current treatments. We explored how galanin receptor 2 (GALR2) and Neuropeptide Y1 Receptor (NPYY1R) agonists, working together, can boost brain cell growth and increase antidepressant-like effects in rats. This suggests new ways to treat Major Depressive Disorder (MDD). RESEARCH DESIGN AND METHODS In a controlled laboratory setting, adult naive Sprague-Dawley rats were administered directly into the brain's ventricles, a method known as intracerebroventricular (ICV) administration, with GALR2 agonist (M1145), NPYY1R agonist, both, or in combination with a GALR2 antagonist (M871). Main outcome measures included long-term neuronal survival, differentiation, and behavioral. RESULTS Co-administration of M1145 and NPYY1R agonist significantly enhanced neuronal survival and maturation in the ventral dentate gyrus, with a notable increase in Brain-Derived Neurotrophic Factor (BDNF) expression. This neurogenic effect was associated with an antidepressant-like effect, an outcome partially reversed by M871. CONCLUSIONS GALR2 and NPYY1R agonists jointly promote hippocampal neurogenesis and exert antidepressant-like effects in rats without adverse outcomes, highlighting their therapeutic potential for MDD. The study's reliance on an animal model and intracerebroventricular delivery warrants further clinical exploration to confirm these promising results.
Collapse
MESH Headings
- Animals
- Male
- Rats
- Antidepressive Agents/pharmacology
- Antidepressive Agents/administration & dosage
- Brain-Derived Neurotrophic Factor/metabolism
- Cell Survival/drug effects
- Depressive Disorder, Major/drug therapy
- Depressive Disorder, Major/physiopathology
- Disease Models, Animal
- Neurons/drug effects
- Neurons/metabolism
- Peptides
- Rats, Sprague-Dawley
- Receptor, Galanin, Type 2/metabolism
- Receptors, G-Protein-Coupled
- Receptors, Neuropeptide
- Receptors, Neuropeptide Y/metabolism
- Receptors, Neuropeptide Y/antagonists & inhibitors
Collapse
Affiliation(s)
- Dasiel Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Receptomics and Brain Disorders Lab, Edificio Lopez-Peñalver, Instituto de Investigación Biomédica de Málaga, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
| | - Pedro Serrano-Castro
- Instituto de Investigación Biomédica de Málaga, NeuronLab, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga, Unit of Neurology, Hospital Regional Universitario de Málaga, Málaga, Spain
- Vithas Málaga, Vithas Málaga, Grupo Hospitalario Vithas, Málaga, Spain
| | - Jose Andrés Sánchez-Pérez
- Instituto de Investigación Biomédica de Málaga, NeuronLab, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga, Unit of Psychiatry, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | | | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Manuel Narváez
- Instituto de Investigación Biomédica de Málaga, NeuronLab, Facultad de Medicina, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga, Unit of Neurology, Hospital Regional Universitario de Málaga, Málaga, Spain
- Vithas Málaga, Vithas Málaga, Grupo Hospitalario Vithas, Málaga, Spain
| |
Collapse
|
15
|
Sánchez-Varo R, López-Salas A, Beltran-Casanueva R, Díaz-Sánchez E, Alvarez-Contino JE, Barbancho-Fernández MA, Serrano-Castro P, Fuxe K, Borroto-Escuela DO, García-Casares N, Narváez M. Enhancement of neurogenesis and cognition through intranasal co-delivery of galanin receptor 2 (GALR2) and neuropeptide Y receptor 1 (NPY1R) agonists: a potential pharmacological strategy for cognitive dysfunctions. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2024; 20:6. [PMID: 38549164 PMCID: PMC10976774 DOI: 10.1186/s12993-024-00230-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 02/19/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Spatial memory deficits and reduced neuronal survival contribute to cognitive decline seen in the aging process. Current treatments are limited, emphasizing the need for innovative therapeutic strategies. This research explored the combined effects of intranasally co-administered galanin receptor 2 (GALR2) and neuropeptide Y1 receptor (NPY1R) agonists, recognized for their neural benefits, on spatial memory, neuronal survival, and differentiation in adult rats. After intranasal co-delivery of the GALR2 agonist M1145 and a NPY1R agonist to adult rats, spatial memory was tested with the object-in-place task 3 weeks later. We examined neuronal survival and differentiation by assessing BrdU-IR profiles and doublecortin (DCX) labeled cells, respectively. We also used the GALR2 antagonist M871 to confirm GALR2's crucial role in promoting cell growth. RESULTS Co-administration improved spatial memory and increased the survival rate of mature neurons. The positive effect of GALR2 in cell proliferation was confirmed by the nullifying effects of its antagonist. The treatment boosted DCX-labeled newborn neurons and altered dendritic morphology, increasing cells with mature dendrites. CONCLUSIONS Our results show that intranasal co-delivery of GALR2 and NPY1R agonists improves spatial memory, boosts neuronal survival, and influences neuronal differentiation in adult rats. The significant role of GALR2 is emphasized, suggesting new potential therapeutic strategies for cognitive decline.
Collapse
Affiliation(s)
- Raquel Sánchez-Varo
- NeuronLab. Departamento Fisiología Humana, Histología Humana, Anatomía Patológica y Educación Física y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071, Malaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA-Plataforma Bionand, Universidad de Malaga, 29071, Malaga, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Alexander López-Salas
- NeuronLab. Departamento Fisiología Humana, Histología Humana, Anatomía Patológica y Educación Física y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071, Malaga, Spain
- Receptomics and Brain Disorders Lab, Universidad de Málaga. Facultad de Medicina., Edificio Lopez-Peñalver, Jimenez Fraud 10, 29071, Málaga, Spain
| | - Rasiel Beltran-Casanueva
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Receptomics and Brain Disorders Lab, Universidad de Málaga. Facultad de Medicina., Edificio Lopez-Peñalver, Jimenez Fraud 10, 29071, Málaga, Spain
| | - Estela Díaz-Sánchez
- NeuronLab. Departamento Fisiología Humana, Histología Humana, Anatomía Patológica y Educación Física y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071, Malaga, Spain
- Vithas Málaga., Vithas Málaga. Grupo Hospitalario Vithas, Málaga, Spain
| | - Jose Erik Alvarez-Contino
- NeuronLab. Departamento Fisiología Humana, Histología Humana, Anatomía Patológica y Educación Física y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071, Malaga, Spain
- Receptomics and Brain Disorders Lab, Universidad de Málaga. Facultad de Medicina., Edificio Lopez-Peñalver, Jimenez Fraud 10, 29071, Málaga, Spain
| | - Miguel Angel Barbancho-Fernández
- NeuronLab. Departamento Fisiología Humana, Histología Humana, Anatomía Patológica y Educación Física y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071, Malaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA-Plataforma Bionand, Universidad de Malaga, 29071, Malaga, Spain
| | - Pedro Serrano-Castro
- Instituto de Investigación Biomédica de Málaga-IBIMA-Plataforma Bionand, Universidad de Malaga, 29071, Malaga, Spain
- Vithas Málaga., Vithas Málaga. Grupo Hospitalario Vithas, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga, Unit of Neurology, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Dasiel O Borroto-Escuela
- NeuronLab. Departamento Fisiología Humana, Histología Humana, Anatomía Patológica y Educación Física y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071, Malaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA-Plataforma Bionand, Universidad de Malaga, 29071, Malaga, Spain
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Receptomics and Brain Disorders Lab, Universidad de Málaga. Facultad de Medicina., Edificio Lopez-Peñalver, Jimenez Fraud 10, 29071, Málaga, Spain
| | - Natalia García-Casares
- NeuronLab. Departamento Fisiología Humana, Histología Humana, Anatomía Patológica y Educación Física y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071, Malaga, Spain
- Instituto de Investigación Biomédica de Málaga-IBIMA-Plataforma Bionand, Universidad de Malaga, 29071, Malaga, Spain
- Departamento de Medicina y Dermatología. , Facultad de Medicina. Universidad de Málaga. , Málaga, Spain
| | - Manuel Narváez
- NeuronLab. Departamento Fisiología Humana, Histología Humana, Anatomía Patológica y Educación Física y Deportiva, Facultad de Medicina, Universidad de Malaga, 29071, Malaga, Spain.
- Instituto de Investigación Biomédica de Málaga-IBIMA-Plataforma Bionand, Universidad de Malaga, 29071, Malaga, Spain.
- Vithas Málaga., Vithas Málaga. Grupo Hospitalario Vithas, Málaga, Spain.
| |
Collapse
|
16
|
Mayne P, Das J, Zou S, Sullivan RKP, Burne THJ. Perineuronal nets are associated with decision making under conditions of uncertainty in female but not male mice. Behav Brain Res 2024; 461:114845. [PMID: 38184206 DOI: 10.1016/j.bbr.2024.114845] [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: 10/16/2023] [Revised: 12/21/2023] [Accepted: 01/02/2024] [Indexed: 01/08/2024]
Abstract
Biological sex influences decision-making processes in significant ways, differentiating the responses animals choose when faced with a range of stimuli. The neurobiological underpinnings that dictate sex differences in decision-making tasks remains an important open question, yet single-sex studies of males form most studies in behavioural neuroscience. Here we used female and male BALB/c mice on two spatial learning and memory tasks and examined the expression of perineuronal nets (PNNs) and parvalbumin interneurons (PV) in regions correlated with spatial memory. Mice underwent the aversive active place avoidance (APA) task or the appetitive trial-unique nonmatching-to-location (TUNL) touchscreen task. Mice in the APA cohort learnt to avoid the foot-shock and no differences were observed on key measures of the task nor in the number and intensity of PNNs and PV. On the delay but not separation manipulation in the TUNL task, females received more incorrect trials and less correct trials compared to males. Furthermore, females in this cohort exhibited higher intensity PNNs and PV cells in the agranular and granular retrosplenial cortex, compared to males. These data show that female and male mice perform similarly on spatial learning tasks. However, sex differences in neural circuitry may underly differences in making decisions under conditions of uncertainty on an appetitive task. These data emphasise the importance of using mice of both sexes in studies of decision-making neuroscience.
Collapse
Affiliation(s)
- Phoebe Mayne
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Joyosmita Das
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Simin Zou
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Robert K P Sullivan
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Thomas H J Burne
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia; Queensland Centre for Mental Health Research, Wacol, QLD 4076, Australia.
| |
Collapse
|
17
|
Velikic G, Maric DM, Maric DL, Supic G, Puletic M, Dulic O, Vojvodic D. Harnessing the Stem Cell Niche in Regenerative Medicine: Innovative Avenue to Combat Neurodegenerative Diseases. Int J Mol Sci 2024; 25:993. [PMID: 38256066 PMCID: PMC10816024 DOI: 10.3390/ijms25020993] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/30/2023] [Accepted: 12/06/2023] [Indexed: 01/24/2024] Open
Abstract
Regenerative medicine harnesses the body's innate capacity for self-repair to restore malfunctioning tissues and organs. Stem cell therapies represent a key regenerative strategy, but to effectively harness their potential necessitates a nuanced understanding of the stem cell niche. This specialized microenvironment regulates critical stem cell behaviors including quiescence, activation, differentiation, and homing. Emerging research reveals that dysfunction within endogenous neural stem cell niches contributes to neurodegenerative pathologies and impedes regeneration. Strategies such as modifying signaling pathways, or epigenetic interventions to restore niche homeostasis and signaling, hold promise for revitalizing neurogenesis and neural repair in diseases like Alzheimer's and Parkinson's. Comparative studies of highly regenerative species provide evolutionary clues into niche-mediated renewal mechanisms. Leveraging endogenous bioelectric cues and crosstalk between gut, brain, and vascular niches further illuminates promising therapeutic opportunities. Emerging techniques like single-cell transcriptomics, organoids, microfluidics, artificial intelligence, in silico modeling, and transdifferentiation will continue to unravel niche complexity. By providing a comprehensive synthesis integrating diverse views on niche components, developmental transitions, and dynamics, this review unveils new layers of complexity integral to niche behavior and function, which unveil novel prospects to modulate niche function and provide revolutionary treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Gordana Velikic
- Department for Research and Development, Clinic Orto MD-Parks Dr. Dragi Hospital, 21000 Novi Sad, Serbia
- Hajim School of Engineering, University of Rochester, Rochester, NY 14627, USA
| | - Dusan M. Maric
- Department for Research and Development, Clinic Orto MD-Parks Dr. Dragi Hospital, 21000 Novi Sad, Serbia
- Faculty of Stomatology Pancevo, University Business Academy, 26000 Pancevo, Serbia;
| | - Dusica L. Maric
- Department of Anatomy, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Gordana Supic
- Institute for Medical Research, Military Medical Academy, 11000 Belgrade, Serbia; (G.S.); (D.V.)
- Medical Faculty of Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
| | - Miljan Puletic
- Faculty of Stomatology Pancevo, University Business Academy, 26000 Pancevo, Serbia;
| | - Oliver Dulic
- Department of Surgery, Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia;
| | - Danilo Vojvodic
- Institute for Medical Research, Military Medical Academy, 11000 Belgrade, Serbia; (G.S.); (D.V.)
- Medical Faculty of Military Medical Academy, University of Defense, 11000 Belgrade, Serbia
| |
Collapse
|
18
|
Chintamen S, Gaur P, Vo N, Bradshaw EM, Menon V, Kernie SG. Distinct microglial transcriptomic signatures within the hippocampus. PLoS One 2024; 19:e0296280. [PMID: 38180982 PMCID: PMC10775894 DOI: 10.1371/journal.pone.0296280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 12/08/2023] [Indexed: 01/07/2024] Open
Abstract
Microglia, the resident immune cells of the brain, are crucial in the development of the nervous system. Recent evidence demonstrates that microglia modulate adult hippocampal neurogenesis by inhibiting cell proliferation of neural precursors and survival both in vitro and in vivo, thus maintaining a balance between cell division and cell death in the neural stem cell pool. There are increasing reports suggesting these microglia found in neurogenic niches differ from their counterparts in non-neurogenic areas. Here, we present evidence that hippocampal microglia exhibit transcriptomic heterogeneity, with some cells expressing genes associated with neurogenesis. By comprehensively profiling myeloid lineage cells in the hippocampus using single cell RNA-sequencing, we have uncovered a small, yet distinct population of microglia which exhibit depletion in genes associated with homeostatic microglia and enrichment of genes associated with phagocytosis. Intriguingly, this population also expresses a gene signature with substantial overlap with previously characterized phenotypes, including disease associated microglia (DAM), a particularly unique and compelling microglial state.
Collapse
Affiliation(s)
- Sana Chintamen
- Department of Pediatrics, Columbia University College of Physicians and
Surgeons, New York, New York, United States of America
| | - Pallavi Gaur
- Department of Neurology, Columbia University College of Physicians and
Surgeons, New York, New York, United States of America
| | - Nicole Vo
- Department of Neurology, Columbia University College of Physicians and
Surgeons, New York, New York, United States of America
| | - Elizabeth M. Bradshaw
- Department of Neurology, Columbia University College of Physicians and
Surgeons, New York, New York, United States of America
| | - Vilas Menon
- Department of Neurology, Columbia University College of Physicians and
Surgeons, New York, New York, United States of America
| | - Steven G. Kernie
- Department of Pediatrics, Columbia University College of Physicians and
Surgeons, New York, New York, United States of America
- Department of Neurology, Columbia University College of Physicians and
Surgeons, New York, New York, United States of America
| |
Collapse
|
19
|
Newell AJ, Patisaul HB. Developmental organophosphate flame retardant exposure disrupts adult hippocampal neurogenesis in Wistar rats. Neurotoxicology 2023; 99:104-114. [PMID: 37783313 PMCID: PMC10842265 DOI: 10.1016/j.neuro.2023.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/04/2023]
Abstract
Organophosphate flame retardant (OPFR) contamination is ubiquitous and bio-monitoring studies have shown that human exposure is widespread and may be unavoidable. OPFRs bear structural similarities to known neurotoxicants such as organophosphate insecticides and have been shown to have both endocrine disrupting and developmental neurotoxic effects. The perinatal period in rodents represents a critical period in the organization of the developing nervous system and insults during this time can impart profound changes on the trajectory of neural development and function, lasting into adulthood. Adult hippocampal neurogenesis (AHN) facilitates dentate gyrus function and broader hippocampal circuit activity in adults; however, the neurogenic potential of this process in adulthood is vulnerable to disruption by exogenous factors during early life. We sought to assess the impact of OPFRs on AHN in offspring of dams exposed during gestation and lactation. Results indicate that developmental OPFR exposure has significant, sex specific impacts on multiple markers of AHN in the dentate gyrus of rats. In males, OPFR exposure significantly reduced the number of neural progenitors the number of new/immature neurons and reduced dentate gyrus volume. In females, exposure increased the number of neural progenitors, decreased the number of new/immature neurons, but had no significant effect on dentate gyrus volume. These results further elucidate the developmental neurotoxic properties of OPFRs, emphasize the long-term impact of early life OPFR exposure on neural processes, and highlight the importance of including sex as a biological variable in neurotoxicology research.
Collapse
Affiliation(s)
- Andrew J Newell
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA.
| | - Heather B Patisaul
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
20
|
Monari PK, Herro ZJ, Bymers J, Marler CA. Chronic intranasal oxytocin increases acoustic eavesdropping and adult neurogenesis. Horm Behav 2023; 156:105443. [PMID: 37871536 DOI: 10.1016/j.yhbeh.2023.105443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 10/25/2023]
Abstract
Social information gathering is a complex process influenced by neuroendocrine-modulated neural plasticity. Oxytocin (OXT) is a key regulator of social decision-making processes such as information gathering, as it contextually modulates social salience and can induce long-term structural plasticity, including neurogenesis. Understanding the link between OXT-induced plasticity and communicative awareness is crucial, particularly because OXT is being considered for treatment of social pathologies. We investigated the role of chronic OXT-dependent plasticity in attention to novel social information by manipulating the duration of time following cessation of intranasal treatment to allow for the functional integration of adult-born neurons resulting from OXT treatment. Following a 3-week delay, chronic intranasal OXT (IN-OXT) increased approach behavior of both female and male mice towards aggressive vocal playbacks of two unseen novel conspecifics, while no effect was observed after a 3-day delay. Immature neurons increased in the ventral hippocampus of females and males treated with chronic IN-OXT after the 3-week delay, indicating a potential association between ventral hippocampal neurogenesis and approach/acoustic eavesdropping. The less the mouse approached, the higher the level of neurogenesis. Contrary to expectations, the correlation between ventral hippocampal neurogenesis and approach behavior was not affected by IN-OXT, suggesting that other plasticity mechanisms underlie the long-term effects of chronic OXT on social approach. Furthermore, we found a negative correlation between ventral hippocampal neurogenesis and freezing behavior. Overall, our results demonstrate that chronic IN-OXT-induced long-term plasticity can influence approach to vocal information and we further reinforced the link between neurogenesis and anxiety.
Collapse
Affiliation(s)
- Patrick K Monari
- Department of Psychology, University of Wisconsin-Madison, WI, USA.
| | - Zachary J Herro
- Department of Psychology, University of Wisconsin-Madison, WI, USA
| | - Jessica Bymers
- Department of Psychology, University of Wisconsin-Madison, WI, USA
| | | |
Collapse
|
21
|
Chandwani MN, Kamte YS, Singh VR, Hemerson ME, Michaels AC, Leak RK, O'Donnell LA. The anti-viral immune response of the adult host robustly modulates neural stem cell activity in spatial, temporal, and sex-specific manners. Brain Behav Immun 2023; 114:61-77. [PMID: 37516388 DOI: 10.1016/j.bbi.2023.07.008] [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: 02/23/2023] [Revised: 06/20/2023] [Accepted: 07/14/2023] [Indexed: 07/31/2023] Open
Abstract
Viruses induce a wide range of neurological sequelae through the dysfunction and death of infected cells and persistent inflammation in the brain. Neural stem cells (NSCs) are often disturbed during viral infections. Although some viruses directly infect and kill NSCs, the antiviral immune response may also indirectly affect NSCs. To better understand how NSCs are influenced by a productive immune response, where the virus is successfully resolved and the host survives, we used the CD46+ mouse model of neuron-restricted measles virus (MeV) infection. As NSCs are spared from direct infection in this model, they serve as bystanders to the antiviral immune response initiated by selective infection of mature neurons. MeV-infected mice showed distinct regional and temporal changes in NSCs in the primary neurogenic niches of the brain, the hippocampus and subventricular zone (SVZ). Hippocampal NSCs increased throughout the infection (7 and 60 days post-infection; dpi), while mature neurons transiently declined at 7 dpi and then rebounded to basal levels by 60 dpi. In the SVZ, NSC numbers were unchanged, but mature neurons declined even after the infection was controlled at 60 dpi. Further analyses demonstrated sex, temporal, and region-specific changes in NSC proliferation and neurogenesis throughout the infection. A relatively long-term increase in NSC proliferation and neurogenesis was observed in the hippocampus; however, neurogenesis was reduced in the SVZ. This decline in SVZ neurogenesis was associated with increased immature neurons in the olfactory bulb in female, but not male mice, suggesting potential migration of newly-made neurons out of the female SVZ. These sex differences in SVZ neurogenesis were accompanied by higher infiltration of B cells and greater expression of interferon-gamma and interleukin-6 in female mice. Learning, memory, and olfaction tests revealed no overt behavioral changes after the acute infection subsided. These results indicate that antiviral immunity modulates NSC activity in adult mice without inducing gross behavioral deficits among those tested, suggestive of mechanisms to restore neurons and maintain adaptive behavior, but also revealing the potential for robust NSC disruption in subclinical infections.
Collapse
Affiliation(s)
- Manisha N Chandwani
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA
| | - Yashika S Kamte
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA
| | - Vivek R Singh
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA
| | - Marlo E Hemerson
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA
| | - Alexa C Michaels
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA
| | - Rehana K Leak
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA
| | - Lauren A O'Donnell
- Duquesne University School of Pharmacy, Graduate School of Pharmaceutical Sciences, Pittsburgh, PA, USA.
| |
Collapse
|
22
|
Garcia-Segura LM, Méndez P, Arevalo MA, Azcoitia I. Neuroestradiol and neuronal development: Not an exclusive male tale anymore. Front Neuroendocrinol 2023; 71:101102. [PMID: 37689249 DOI: 10.1016/j.yfrne.2023.101102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
The brain synthesizes a variety of neurosteroids, including neuroestradiol. Inhibition of neuroestradiol synthesis results in alterations in basic neurodevelopmental processes, such as neurogenesis, neuroblast migration, neuritogenesis and synaptogenesis. Although the neurodevelopmental actions of neuroestradiol are exerted in both sexes, some of them are sex-specific, such as the well characterized effects of neuroestradiol derived from the metabolism of testicular testosterone during critical periods of male brain development. In addition, recent findings have shown sex-specific actions of neuroestradiol on neuroblast migration, neuritic growth and synaptogenesis in females. Among other factors, the epigenetic regulation exerted by X linked genes, such as Kdm6a/Utx, may determine sex-specific actions of neuroestradiol in the female brain. This review evidences the impact of neuroestradiol on brain formation in both sexes and highlights the interaction of neural steriodogenesis, hormones and sex chromosomes in sex-specific brain development.
Collapse
Affiliation(s)
- Luis M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto Nacional de Salud Carlos III, Madrid, Spain.
| | - Pablo Méndez
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain
| | - M Angeles Arevalo
- Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto Nacional de Salud Carlos III, Madrid, Spain.
| | - Iñigo Azcoitia
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto Nacional de Salud Carlos III, Madrid, Spain; Department of Cell Biology, Universidad Complutense de Madrid, C José Antonio Nováis 12, 28040 Madrid, Spain
| |
Collapse
|
23
|
Colom-Rocha C, Bis-Humbert C, García-Fuster MJ. Evaluating signs of hippocampal neurotoxicity induced by a revisited paradigm of voluntary ethanol consumption in adult male and female Sprague-Dawley rats. Pharmacol Rep 2023; 75:320-330. [PMID: 36807777 DOI: 10.1007/s43440-023-00464-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUND Binge alcohol drinking is considered a prominent risk factor for the development of alcohol-use disorders, and could be model in rodents through the standard two-bottle preference choice test. The goal was to recreate an intermittent use of alcohol during 3 consecutive days each week to ascertain its potential impact on hippocampal neurotoxicity (neurogenesis and other neuroplasticity markers), and including sex as a biological variable, given the well-known sex differences in alcohol consumption. METHODS Ethanol access was granted to adult Sprague-Dawley rats for 3 consecutive days per week, followed by 4 days of withdrawal, during 6 weeks, mimicking the most common pattern of intake in people, drinking over the weekends in an intensive manner. Hippocampal samples were collected to evaluate signs of neurotoxicity. RESULTS Female rats consumed significantly more ethanol than males, although intake did not escalate over time. Ethanol preference levels remained below 40% over time and did not differ between sexes. Moderate signs of ethanol neurotoxicity were observed in hippocampus at the level of decreased neuronal progenitors (NeuroD + cells), and these effects were independent of sex. No other signs of neurotoxicity were induced by ethanol voluntary consumption when measured through several key cell fate markers (i.e., FADD, Cyt c, Cdk5, NF-L) by western blot analysis. CONCLUSIONS Overall, the present results suggest that even though we modeled a situation where no escalation in ethanol intake occurred across time, mild signs of neurotoxicity emerged, suggesting that even the use of ethanol during adulthood in a recreational way could lead to certain brain harm.
Collapse
Affiliation(s)
- Carles Colom-Rocha
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa Km 7.5, 07122, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
| | - Cristian Bis-Humbert
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa Km 7.5, 07122, Palma, Spain.,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.,Psychobiology of Drug Addiction, Neurocentre Magendie, INSERM U1215, Bordeaux, France
| | - M Julia García-Fuster
- IUNICS, University of the Balearic Islands, Cra. de Valldemossa Km 7.5, 07122, Palma, Spain. .,Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain.
| |
Collapse
|
24
|
Local CRF and oxytocin receptors correlate with female experience-driven avoidance change and hippocampal neuronal plasticity. Neurochem Int 2023; 163:105485. [PMID: 36623734 DOI: 10.1016/j.neuint.2023.105485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 12/12/2022] [Accepted: 01/05/2023] [Indexed: 01/09/2023]
Abstract
Understanding how experiences affect females' behaviors and neuronal plasticity is essential for uncovering the mechanism of neurodevelopmental disorders. The study explored how neonatal maternal deprivation (MD) and post-weaning environmental enrichment (EE) impacted the CA1 and DG's neuronal plasticity in the dorsal hippocampus, and its relationships with passive avoidance, local corticotrophin-releasing factor (CRF) levels, and oxytocin receptor (OTR) levels in female BALB/c mice. The results showed that MD damaged passive avoidance induced by foot shock and hotness, and EE restored it partially. In the CA1, MD raised CRF levels and OTR levels. Parallelly, MD increased synaptic connection levels but reduced the branches' numbers of pyramidal neurons. Meanwhile, in the DG, MD increased OTR levels but lowered CRF levels, DNA levels, and spine densities. EE did not change the CA1 and DG's CRF and OTR levels. However, EE added DG's dendrites of granular cells. The additive of MD and EE raised CA1's synaptophysin and DG's postsynaptic density protein-95 and OTR levels, and meanwhile, shaped avoidance behaviors primarily similar to the control. The results suggest that experience-driven avoidance change and hippocampal neuronal plasticity are associated with local CRF and OTR levels in female mice.
Collapse
|
25
|
Zhang TR, Askari B, Kesici A, Guilherme E, Vila-Rodriguez F, Snyder JS. Intermittent theta burst transcranial magnetic stimulation induces hippocampal mossy fibre plasticity in male but not female mice. Eur J Neurosci 2023; 57:310-323. [PMID: 36484786 DOI: 10.1111/ejn.15891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 11/12/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022]
Abstract
Transcranial magnetic stimulation (TMS) induces electric fields that depolarise or hyperpolarise neurons. Intermittent theta burst stimulation (iTBS), a patterned form of TMS that is delivered at the theta frequency (~5 Hz), induces neuroplasticity in the hippocampus, a brain region that is implicated in memory and learning. One form of plasticity that is unique to the hippocampus is adult neurogenesis; however, little is known about whether TMS or iTBS in particular affects newborn neurons. Here, we therefore applied repeated sessions of iTBS to male and female mice and measured the extent of adult neurogenesis and the morphological features of immature neurons. We found that repeated sessions of iTBS did not significantly increase the amount of neurogenesis or affect the gross dendritic morphology of new neurons, and there were no sex differences in neurogenesis rates or aspects of afferent morphology. In contrast, efferent properties of newborn neurons varied as a function of sex and stimulation. Chronic iTBS increased the size of mossy fibre terminals, which synapse onto Cornu Ammonis 3 (CA3) pyramidal neurons, but only in males. iTBS also increased the number of terminal-associated filopodia, putative synapses onto inhibitory interneurons but only in male mice. This efferent plasticity could result from a general trophic effect, or it could reflect accelerated maturation of immature neurons. Given the important role of mossy fibre synapses in hippocampal learning, our results identify a neurobiological effect of iTBS that might be associated with sex-specific changes in cognition.
Collapse
Affiliation(s)
- Tian Rui Zhang
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Non-Invasive Neurostimulation Therapies Laboratory, Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Baran Askari
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Aydan Kesici
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Evelyn Guilherme
- Department of Physiotherapy, Federal University of Sao Carlos, Sao Carlo, Brazil
| | - Fidel Vila-Rodriguez
- Non-Invasive Neurostimulation Therapies Laboratory, Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jason S Snyder
- Department of Psychology, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
26
|
Galea LA, Lee BH, de leon RG, Rajah MN, Einstein G. Beyond sex and gender differences: The case for women's health research. PRINCIPLES OF GENDER-SPECIFIC MEDICINE 2023:699-711. [DOI: 10.1016/b978-0-323-88534-8.00045-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
27
|
Akinbo OI, McNeal N, Hylin M, Hite N, Dagner A, Grippo AJ. The Influence of Environmental Enrichment on Affective and Neural Consequences of Social Isolation Across Development. AFFECTIVE SCIENCE 2022; 3:713-733. [PMID: 36519141 PMCID: PMC9743881 DOI: 10.1007/s42761-022-00131-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 06/10/2022] [Indexed: 05/15/2023]
Abstract
Social stress is associated with depression and anxiety, physiological disruptions, and altered brain morphology in central stress circuitry across development. Environmental enrichment strategies may improve responses to social stress. Socially monogamous prairie voles exhibit analogous social and emotion-related behaviors to humans, with potential translational insight into interactions of social stress, age, and environmental enrichment. This study explored the effects of social isolation and environmental enrichment on behaviors related to depression and anxiety, physiological indicators of stress, and dendritic structural changes in amygdala and hippocampal subregions in young adult and aging prairie voles. Forty-nine male prairie voles were assigned to one of six groups divided by age (young adult vs. aging), social structure (paired vs. isolated), and housing environment (enriched vs. non-enriched). Following 4 weeks of these conditions, behaviors related to depression and anxiety were investigated in the forced swim test and elevated plus maze, body and adrenal weights were evaluated, and dendritic morphology analyses were conducted in hippocampus and amygdala subregions. Environmental enrichment decreased immobility duration in the forced swim test, increased open arm exploration in the elevated plus maze, and reduced adrenal/body weight ratio in aging and young adult prairie voles. Age and social isolation influenced dendritic morphology in the basolateral amygdala. Age, but not social isolation, influenced dendritic morphology in the hippocampal dentate gyrus. Environmental enrichment did not influence dendritic morphology in either brain region. These data may inform interventions to reduce the effects of social stressors and age-related central changes associated with affective behavioral consequences in humans.
Collapse
Affiliation(s)
- Oreoluwa I. Akinbo
- Department of Psychology, Northern Illinois University, DeKalb, IL 60115 USA
| | - Neal McNeal
- Department of Psychology, Northern Illinois University, DeKalb, IL 60115 USA
| | - Michael Hylin
- Department of Psychology, Southern Illinois University, Carbondale, IL 62901 USA
| | - Natalee Hite
- Department of Physiology, Southern Illinois University, Carbondale, IL, 62901, USA
| | - Ashley Dagner
- Department of Psychology, Northern Illinois University, DeKalb, IL 60115 USA
| | - Angela J. Grippo
- Department of Psychology, Northern Illinois University, DeKalb, IL 60115 USA
| |
Collapse
|
28
|
Yagi S, Lee A, Truter N, Galea LAM. Sex differences in contextual pattern separation, neurogenesis, and functional connectivity within the limbic system. Biol Sex Differ 2022; 13:42. [PMID: 35870952 PMCID: PMC9308289 DOI: 10.1186/s13293-022-00450-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/05/2022] [Indexed: 01/04/2023] Open
Abstract
Background Females are more likely to present with anxiety disorders such as post-traumatic stress disorder (PTSD) compared to males, which are associated with disrupted hippocampal integrity. Sex differences in the structure and function of hippocampus exist. Here, we examined sex differences in contextual pattern separation, functional connectivity, and activation of new neurons during fear memory. Methods Two-month-old male and female Sprague-Dawley rats were injected with the DNA synthesis markers, iododeoxyuridine (IdU) and chlorodeoxyuridine (CldU) 3 weeks and 4 weeks before perfusion, respectively. One week after CldU injection, the rats underwent a context discrimination task in which rats were placed in context A (shock) and context A’ (no shock) every day for 12 days. On the test day, rats were placed in the shock context (context A) to measure fear memory and expression of zif268, an immediate early gene across 16 different limbic and reward regions. Repeated-measures or factorial analysis of variance was conducted on our variables of interest. Pearson product-moment calculations and principal component analyses on zif268 expression across regions were also performed. Results We found that females, but not males, showed contextual discrimination during the last days of training. On the test day, both sexes displayed similar levels of freezing, indicating equivalent fear memory for context A. Despite similar fear memory, males showed more positive correlations of zif268 activation between the limbic regions and the striatum, whereas females showed more negative correlations among these regions. Females showed greater activation of the frontal cortex, dorsal CA1, and 3-week-old adult-born dentate granular cells compared to males. Conclusions These results highlight the importance of studying sex differences in fear memory and the contribution of adult neurogenesis to the neuronal network and may contribute to differences in susceptibility to fear-related disorders such as post-traumatic stress disorder. HighlightsFemale rats, but not male rats, show faster discrimination during a contextual pattern separation task. Three-week-old adult-born neurons are more active in response to fear memory in females compared to males. Females had greater neural activation compared to males in the frontal cortex and dorsal CA1 region of the hippocampus in response to fear memory. Males and females show distinct patterns in functional connectivity for fear memory across limbic regions. Males have many positive correlations between activated new neurons of different ages between the dorsal and ventral hippocampus, while females show more correlations between activated new neurons and other limbic regions.
Supplementary Information The online version contains supplementary material available at 10.1186/s13293-022-00450-2.
Collapse
|
29
|
An analysis of neuroscience and psychiatry papers published from 2009 and 2019 outlines opportunities for increasing discovery of sex differences. Nat Commun 2022; 13:2137. [PMID: 35440664 PMCID: PMC9018784 DOI: 10.1038/s41467-022-29903-3] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 04/07/2022] [Indexed: 12/12/2022] Open
Abstract
Sex differences exist in many neurological and psychiatric diseases, but these have not always been addressed adequately in research. In order to address this, it is necessary to consider how sex is incorporated into the design (e.g. using a balanced design) and into the analyses (e.g. using sex as a covariate) in the published literature. We surveyed papers published in 2009 and 2019 across six journals in neuroscience and psychiatry. In this sample, we find a 30% increase in the percentage of papers reporting studies that included both sexes in 2019 compared with 2009. Despite this increase, in 2019 only 19% of papers in the sample reported using an optimal design for discovery of possible sex differences, and only 5% of the papers reported studies that analysed sex as a discovery variable. We conclude that progress to date has not been sufficient to address the importance of sex differences in research for discovery and therapeutic potential for neurological and psychiatric disease. Sex differences occur in many neurological and psychiatric diseases, and yet research is not always designed optimally to identify these. Here the authors perform a study of how sex was incorporated into the design and analyses of papers published six journals in neuroscience and psychiatry in 2009 compared with 2019.
Collapse
|
30
|
Corticosterone induces discrete epigenetic signatures in the dorsal and ventral hippocampus that depend upon sex and genotype: focus on methylated Nr3c1 gene. Transl Psychiatry 2022; 12:109. [PMID: 35296634 PMCID: PMC8927334 DOI: 10.1038/s41398-022-01864-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/17/2022] [Accepted: 02/24/2022] [Indexed: 02/07/2023] Open
Abstract
The genomic effects of circulating glucocorticoids are particularly relevant in cortico-limbic structures, which express a high concentration of steroid hormone receptors. To date, no studies have investigated genomic differences in hippocampal subregions, namely the dorsal (dHPC) and ventral (vHPC) hippocampus, in preclinical models treated with exogenous glucocorticoids. Chronic oral corticosterone (CORT) in mouse is a pharmacological approach that disrupts the activity of the hypothalamic-pituitary-adrenal axis, increases affective behavior, and induces genomic changes after stress in the HPC of wildtype (WT) mice and mice heterozygous for the gene coding for brain-derived neurotrophic factor Val66Met (hMet), a variant associated with genetic susceptibility to stress. Using RNA-sequencing, we investigated the genomic signatures of oral CORT in the dHPC and vHPC of WT and hMet male and female mice, and examined sex and genotype differences in response to oral CORT. Males under CORT showed lower glycemia and increased anxiety- and depression-like behavior compared to females that showed instead opposite affective behavior in response to CORT. Rank-rank-hypergeometric overlap (RRHO) was used to identify genes from a continuous gradient of significancy that were concordant across groups. RRHO showed that CORT-induced differentially expressed genes (DEGs) in WT mice and hMet mice converged in the dHPC of males and females, while in the vHPC, DEGs converged in males and diverged in females. The vHPC showed a higher number of DEGs compared to the dHPC and exhibited sex differences related to glucocorticoid receptor (GR)-binding genes and epigenetic modifiers. Methyl-DNA-immunoprecipitation in the vHPC revealed differential methylation of the exons 1C and 1F of the GR gene (Nr3c1) in hMet females. Together, we report behavioral and endocrinological sex differences in response to CORT, as well as epigenetic signatures that i) differ in the dHPC and vHPC,ii) are distinct in males and females, and iii) implicate differential methylation of Nr3c1 selectively in hMet females.
Collapse
|
31
|
Cabrera-Muñoz EA, Olvera-Hernández S, Vega-Rivera NM, Meneses-San Juan D, Reyes-Haro D, Ortiz-López L, Ramírez Rodríguez GB. Environmental Enrichment Differentially Activates Neural Circuits in FVB/N Mice, Inducing Social Interaction in Females but Agonistic Behavior in Males. Neurochem Res 2022; 47:781-794. [PMID: 34978003 DOI: 10.1007/s11064-021-03487-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/14/2021] [Accepted: 11/09/2021] [Indexed: 01/17/2023]
Abstract
Environmental enrichment induces behavioral and structural modifications in rodents and influences the capability of mice to cope with stress. However, little is understood about hippocampal neurogenesis and the appearance of social/agonistic (aggressive) behavior upon activation of different neuronal circuits in FVB/N mice. Thus, in this study we hypothesized that environmental enrichment differentially regulates neurogenesis, neural circuit activation and social/agonistic behavior in male and female FVB/N mice. We explored the (1) neurogenic process as an indicative of neuroplasticity, (2) neuronal activation in the limbic system, and (3) social behavior using the resident-intruder test. On postnatal day 23 (PD23), mice were assigned to one of two groups: Standard Housing or Environmental Enrichment. At PD53, rodents underwent the resident-intruder test to evaluate social behaviors. Results revealed that environmental enrichment increased neurogenesis and social interaction in females. In males, environmental enrichment increased neurogenesis and agonistic behavior. Enriched male mice expressed higher levels of agonistic-related behavior than female mice housed under the same conditions. Neural circuit analysis showed lower activation in the amygdala of enriched males and higher activation in enriched females than their respective controls. Enriched females also showed higher activation in the frontal cortex without differences in male groups. Moreover, the insular cortex was less activated in females than in males. Thus, our results indicate that environmental enrichment has different effects on neuroplasticity and social/agonistic behavior in FVB/N mice, suggesting the relevance of sexual dimorphism in response to environmental stimuli.
Collapse
Affiliation(s)
- Edith Araceli Cabrera-Muñoz
- Laboratorio of Neurogénesis, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría "Ramón de La Fuente Muñiz", Calzada México-Xochimilco No. 101, Colonia San Lorenzo Huipulco, Delegación Tlalpan, C.P. 14370, México City, México
| | - Sandra Olvera-Hernández
- Laboratorio of Neurogénesis, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría "Ramón de La Fuente Muñiz", Calzada México-Xochimilco No. 101, Colonia San Lorenzo Huipulco, Delegación Tlalpan, C.P. 14370, México City, México
| | - Nelly Maritza Vega-Rivera
- Laboratorio of Neuropsicofarmacología, Dirección de Neurociencias, Instituto Nacional de Psiquiatría "Ramón de La Fuente Muñiz", Calzada México-Xochimilco 101, C.P. 14370, México City, México
| | - David Meneses-San Juan
- Laboratorio of Neurogénesis, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría "Ramón de La Fuente Muñiz", Calzada México-Xochimilco No. 101, Colonia San Lorenzo Huipulco, Delegación Tlalpan, C.P. 14370, México City, México
| | - Daniel Reyes-Haro
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología. Universidad Nacional Autónoma de México, Campus Juriquilla. Boulevard Juriquilla 3001, C.P. 76230, Juriquilla, Querétaro, México
| | - Leonardo Ortiz-López
- Laboratorio of Neurogénesis, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría "Ramón de La Fuente Muñiz", Calzada México-Xochimilco No. 101, Colonia San Lorenzo Huipulco, Delegación Tlalpan, C.P. 14370, México City, México
| | - Gerardo Bernabé Ramírez Rodríguez
- Laboratorio of Neurogénesis, Subdirección de Investigaciones Clínicas, Instituto Nacional de Psiquiatría "Ramón de La Fuente Muñiz", Calzada México-Xochimilco No. 101, Colonia San Lorenzo Huipulco, Delegación Tlalpan, C.P. 14370, México City, México.
| |
Collapse
|
32
|
Sheppard PAS, Puri TA, Galea LAM. Sex Differences and Estradiol Effects in MAPK and Akt Cell Signaling across Subregions of the Hippocampus. Neuroendocrinology 2022; 112:621-635. [PMID: 34407537 DOI: 10.1159/000519072] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/16/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Rapid effects of estrogens within the hippocampus of rodents are dependent upon cell-signaling cascades, and activation of these cascades by estrogens varies by sex. Whether these pathways are rapidly activated within the dentate gyrus (DG) and CA1 by estrogens across sex and the anatomical longitudinal axis has been overlooked. METHODS Gonadally intact female and male rats were given either vehicle or physiological systemic low (1.1 µg/kg) or high (37.3 µg/kg) doses of 17β-estradiol 30 min prior to tissue collection. To control for the effects of circulating estrogens, an additional group of female rats was ovariectomized (OVX) and administered 17β-estradiol. Brains were extracted, and tissue punches of the CA1 and DG were taken along the longitudinal hippocampal axis (dorsal and ventral) and analyzed for key mitogen-activated protein kinase (MAPK) and protein kinase B (Akt) cascade phosphoproteins. RESULTS Intact females had higher Akt pathway phosphoproteins (pAkt, pGSK-3β, and pp70S6K) than males in the DG (dorsal and ventral) and lower pERK1/2 in the dorsal DG. Most effects of 17β-estradiol on cell signaling occurred in OVX animals. In OVX animals, 17β-estradiol increased cell signaling of MAPK and Akt phosphoproteins (pERK1/2, pJNK, pAkt, and pGSK-3β) in the CA1 and pERK1/2 and pJNK DG. DISCUSSION/CONCLUSIONS Systemic 17β-estradiol treatment rapidly alters phosphoprotein levels in the hippocampus, dependent on reproductive status, and intact females have greater expression of Akt phosphoproteins than that in intact males in the DG. These findings shed light on underlying mechanisms of sex differences in hippocampal function and response to interventions that affect MAPK or Akt signaling.
Collapse
Affiliation(s)
- Paul A S Sheppard
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tanvi A Puri
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
| | - Liisa A M Galea
- Department of Psychology, University of British Columbia, Vancouver, British Columbia, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
33
|
Harris EP, McGovern AJ, Melo TG, Barron A, Nola YM, O'Leary OF. Juvenile Stress Exerts Sex-independent Effects on Anxiety, Antidepressant-like Behaviours and Dopaminergic Innervation of the Prelimbic Cortex in Adulthood and Does Not Alter Hippocampal Neurogenesis. Behav Brain Res 2021; 421:113725. [PMID: 34929235 DOI: 10.1016/j.bbr.2021.113725] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 11/24/2021] [Accepted: 12/15/2021] [Indexed: 11/26/2022]
Abstract
Stress, particularly during childhood, is a major risk factor for the development of depression. Depression is twice as prevalent in women compared to men, which suggests that that biological sex also contributes to depression susceptibility. However, the neurobiology underpinning sex differences in the long-term consequences of childhood stress remains unknown. Thus, the aim of this study was to determine whether stress applied during the prepubertal juvenile period (postnatal day 27-29) in rats induces sex-specific changes in anxiety-like behaviour, anhedonia, and antidepressant-like behaviour in adulthood in males and females. The impact of juvenile stress on two systems in the brain associated with these behaviours and that develop during the juvenile period, the mesocorticolimbic dopaminergic system and hippocampal neurogenesis, were also investigated. Juvenile stress altered escape-oriented behaviours in the forced swim test in both sexes, decreased latency to drink a palatable substance in a novel environment in the novelty-induced hypophagia test in both sexes, and decreased open field supported rearing behavior in females. These behavioural changes were accompanied by stress-induced increases in tyrosine hydroxylase immunoreactivity in the prefrontal cortex of both sexes, but not other regions of the mesocorticolimbic dopaminergic system. Juvenile stress did not impact anhedonia in adulthood as measured by the saccharin preference test and had no effect hippocampal neurogenesis across the longitudinal axis of the hippocampus. These results suggest that juvenile stress has long-lasting impacts on antidepressant-like and reward-seeking behaviour in adulthood and these changes may be due to alterations to catecholaminergic innervation of the medial prefrontal cortex.
Collapse
Affiliation(s)
- Erin P Harris
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Andrew J McGovern
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Thieza G Melo
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Aaron Barron
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Yvonne M Nola
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Olivia F O'Leary
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Ireland, University College Cork, Ireland.
| |
Collapse
|
34
|
Hodges TE, Puri TA, Blankers SA, Qiu W, Galea LAM. Steroid hormones and hippocampal neurogenesis in the adult mammalian brain. VITAMINS AND HORMONES 2021; 118:129-170. [PMID: 35180925 DOI: 10.1016/bs.vh.2021.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Hippocampal neurogenesis persists across the lifespan in many species, including rodents and humans, and is associated with cognitive performance and the pathogenesis of neurodegenerative disease and psychiatric disorders. Neurogenesis is modulated by steroid hormones that change across development and differ between the sexes in rodents and humans. Here, we discuss the effects of stress and glucocorticoid exposure from gestation to adulthood as well as the effects of androgens and estrogens in adulthood on neurogenesis in the hippocampus. Throughout the review we highlight sex differences in the effects of steroid hormones on neurogenesis and how they may relate to hippocampal function and disease. These data highlight the importance of examining age and sex when evaluating the effects of steroid hormones on hippocampal neurogenesis.
Collapse
Affiliation(s)
- Travis E Hodges
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Tanvi A Puri
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Samantha A Blankers
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Wansu Qiu
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
35
|
Blankers SA, Galea LA. Androgens and Adult Neurogenesis in the Hippocampus. ANDROGENS: CLINICAL RESEARCH AND THERAPEUTICS 2021; 2:203-215. [PMID: 35024692 PMCID: PMC8744005 DOI: 10.1089/andro.2021.0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 08/19/2021] [Indexed: 11/12/2022]
Abstract
Adult neurogenesis in the hippocampus is modulated by steroid hormones, including androgens, in male rodents. In this review, we summarize research showing that chronic exposure to androgens, such as testosterone and dihydrotestosterone, enhances the survival of new neurons in the dentate gyrus of male, but not female, rodents, via the androgen receptor. However, the neurogenesis promoting the effect of androgens in the dentate gyrus may be limited to younger adulthood as it is not evident in middle-aged male rodents. Although direct exposure to androgens in adult or middle age does not significantly influence neurogenesis in female rodents, the aromatase inhibitor letrozole enhances neurogenesis in the hippocampus of middle-aged female mice. Unlike other androgens, androgenic anabolic steroids reduce neurogenesis in the hippocampus of male rodents. Collectively, the research indicates that the ability of androgens to enhance hippocampal neurogenesis in adult rodents is dependent on dose, androgen type, sex, duration, and age. We discuss these findings and how androgens may be influencing neuroprotection, via neurogenesis in the hippocampus, in the context of health and disease.
Collapse
Affiliation(s)
- Samantha A. Blankers
- Graduate Program in Neuroscience, The University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, Canada
| | - Liisa A.M. Galea
- Graduate Program in Neuroscience, The University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, The University of British Columbia, Vancouver, Canada
- Department of Psychology, The University of British Columbia, Vancouver, Canada
| |
Collapse
|
36
|
Nawarawong NN, Thompson KR, Guerin SP, Anasooya Shaji C, Peng H, Nixon K. Reactive, Adult Neurogenesis From Increased Neural Progenitor Cell Proliferation Following Alcohol Dependence in Female Rats. Front Neurosci 2021; 15:689601. [PMID: 34594180 PMCID: PMC8477003 DOI: 10.3389/fnins.2021.689601] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/16/2021] [Indexed: 11/25/2022] Open
Abstract
Hippocampal neurodegeneration is a consequence of excessive alcohol drinking in alcohol use disorders (AUDs), however, recent studies suggest that females may be more susceptible to alcohol-induced brain damage. Adult hippocampal neurogenesis is now well accepted to contribute to hippocampal integrity and is known to be affected by alcohol in humans as well as in animal models of AUDs. In male rats, a reactive increase in adult hippocampal neurogenesis has been observed during abstinence from alcohol dependence, a phenomenon that may underlie recovery of hippocampal structure and function. It is unknown whether reactive neurogenesis occurs in females. Therefore, adult female rats were exposed to a 4-day binge model of alcohol dependence followed by 7 or 14 days of abstinence. Immunohistochemistry (IHC) was used to assess neural progenitor cell (NPC) proliferation (BrdU and Ki67), the percentage of increased NPC activation (Sox2+/Ki67+), the number of immature neurons (NeuroD1), and ectopic dentate gyrus granule cells (Prox1). On day seven of abstinence, ethanol-treated females showed a significant increase in BrdU+ and Ki67+ cells in the subgranular zone of the dentate gyrus (SGZ), as well as greater activation of NPCs (Sox2+/Ki67+) into active cycling. At day 14 of abstinence, there was a significant increase in the number of immature neurons (NeuroD1+) though no evidence of ectopic neurogenesis according to either NeuroD1 or Prox1 immunoreactivity. Altogether, these data suggest that alcohol dependence produces similar reactive increases in NPC proliferation and adult neurogenesis. Thus, reactive, adult neurogenesis may be a means of recovery for the hippocampus after alcohol dependence in females.
Collapse
Affiliation(s)
- Natalie N Nawarawong
- College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - K Ryan Thompson
- College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - Steven P Guerin
- College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | | | - Hui Peng
- Division of Pharmacology & Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| | - Kimberly Nixon
- College of Pharmacy, The University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
37
|
Qiu W, Go KA, Wen Y, Duarte-Guterman P, Eid RS, Galea LAM. Maternal fluoxetine reduces hippocampal inflammation and neurogenesis in adult offspring with sex-specific effects of periadolescent oxytocin. Brain Behav Immun 2021; 97:394-409. [PMID: 34174336 DOI: 10.1016/j.bbi.2021.06.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 06/14/2021] [Accepted: 06/19/2021] [Indexed: 12/22/2022] Open
Abstract
Untreated perinatal depression can have severe consequences for the mother and her children. However, both the efficacy to mothers and safety to exposed infants of pharmacological antidepressants such as selective serotonin reuptake inhibitors (SSRIs), have been questioned. We previously reported that maternal SSRI exposure increased hippocampal IL-1β levels, which may be tied to limited efficacy of SSRIs during the postpartum to the dam but is not yet known whether maternal postpartum SSRIs affect the neuroinflammatory profile of adult offspring. In addition, although controversial, perinatal SSRI exposure has been linked to increased risk of autism spectrum disorder (ASD) in children. Oxytocin (OT) is under investigation as a treatment for ASD, but OT is a large neuropeptide that has difficulty crossing the blood-brain barrier (BBB). TriozanTM is a nanoformulation that can facilitate OT to cross the BBB. Thus, we investigated the impact of maternal postpartum SSRIs and offspring preadolescent OT treatment on adult offspring neuroinflammation, social behavior, and neurogenesis in the hippocampus. Using a model of de novo postpartum depression, corticosterone (CORT) was given in the postpartum to the dam with or without treatment with the SSRI, fluoxetine (FLX) for 21 days postpartum. Offspring were then subsequently treated with either OT, OT + TriozanTM, or vehicle for 10 days prior to adolescence (PD25-34). Maternal FLX decreased hippocampal IL-10 and IL-13 and neurogenesis in both sexes, whereas maternal CORT increased hippocampal IL-13 in both sexes. Maternal CORT treatment shifted the neuroimmune profile towards a more proinflammatory profile in offspring hippocampus, whereas oxytocin, independent of formulation, normalized this profile. OT treatment increased hippocampal neurogenesis in adult males but not in adult females, regardless of maternal treatment. OT treatment increased the time spent with a novel social stimulus animal (social investigation) in both adult male and female offspring, although this effect depended on maternal CORT. These findings underscore that preadolescent exposure to OT can reverse some of the long-lasting effects of postpartum maternal CORT and FLX treatments in the adult offspring. In addition, we found that maternal treatments that reduce (CORT) or increase (FLX) hippocampal inflammation in dams resulted in opposing patterns of hippocampal inflammation in adult offspring.
Collapse
Affiliation(s)
- Wansu Qiu
- Graduate Program in Neuroscience, University of British Columbia, Canada
| | - Kimberly A Go
- Department of Psychology, University of British Columbia, Canada
| | - Yanhua Wen
- Department of Psychology, University of British Columbia, Canada
| | | | - Rand S Eid
- Graduate Program in Neuroscience, University of British Columbia, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Canada; Department of Psychology, University of British Columbia, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada.
| |
Collapse
|
38
|
Wander CM, Song J. The neurogenic niche in Alzheimer's disease. Neurosci Lett 2021; 762:136109. [PMID: 34271133 PMCID: PMC9013442 DOI: 10.1016/j.neulet.2021.136109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 06/17/2021] [Accepted: 07/07/2021] [Indexed: 12/15/2022]
Abstract
Adult hippocampal neurogenesis is the process of generation and functional incorporation of new neurons, formed by adult neural stem cells in the dentate gyrus. Adult hippocampal neurogenesis is highly dependent upon the integration of dynamic external stimuli and is instrumental in the formation of new spatial memories. Adult hippocampal neurogenesis is therefore uniquely sensitive to the summation of neuronal circuit and neuroimmune environments that comprise the neurogenic niche, and has powerful implications in diseases of aging and neurological disorders. This sensitivity underlies the neurogenic niche alterations commonly observed in Alzheimer's disease, the most common form of dementia. This review summarizes Alzheimer's disease associated changes in neuronal network activity, neuroinflammatory processes, and adult neural stem cell fate choice that ultimately result in neurogenic niche dysfunction and impaired adult hippocampal neurogenesis. A more comprehensive understanding of the complex changes mediating neurogenic niche disturbances in Alzheimer's disease will aid development of future therapies targeting adult neurogenesis.
Collapse
Affiliation(s)
- Connor M Wander
- Department of Pharmacology, University of North Carolina at Chapel Hill
| | - Juan Song
- Department of Pharmacology, University of North Carolina at Chapel Hill
- Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
39
|
Bartkowska K, Turlejski K, Tepper B, Rychlik L, Vogel P, Djavadian R. Effects of Brain Size on Adult Neurogenesis in Shrews. Int J Mol Sci 2021; 22:7664. [PMID: 34299282 PMCID: PMC8303847 DOI: 10.3390/ijms22147664] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/14/2021] [Accepted: 07/14/2021] [Indexed: 01/02/2023] Open
Abstract
Shrews are small animals found in many different habitats. Like other mammals, adult neurogenesis occurs in the subventricular zone of the lateral ventricle (SVZ) and the dentate gyrus (DG) of the hippocampal formation. We asked whether the number of new generated cells in shrews depends on their brain size. We examined Crocidura russula and Neomys fodiens, weighing 10-22 g, and Crocidura olivieri and Suncus murinus that weigh three times more. We found that the density of proliferated cells in the SVZ was approximately at the same level in all species. These cells migrated from the SVZ through the rostral migratory stream to the olfactory bulb (OB). In this pathway, a low level of neurogenesis occurred in C. olivieri compared to three other species of shrews. In the DG, the rate of adult neurogenesis was regulated differently. Specifically, the lowest density of newly generated neurons was observed in C. russula, which had a substantial number of new neurons in the OB compared with C. olivieri. We suggest that the number of newly generated neurons in an adult shrew's brain is independent of the brain size, and molecular mechanisms of neurogenesis appeared to be different in two neurogenic structures.
Collapse
Affiliation(s)
- Katarzyna Bartkowska
- Nencki Institute of Experimental Biology Polish Academy of Sciences, 02-093 Warsaw, Poland; (K.B.); (B.T.)
| | - Krzysztof Turlejski
- Faculty of Biology and Environmental Sciences, Cardinal Stefan Wyszynski University in Warsaw, 01-938 Warsaw, Poland;
| | - Beata Tepper
- Nencki Institute of Experimental Biology Polish Academy of Sciences, 02-093 Warsaw, Poland; (K.B.); (B.T.)
| | - Leszek Rychlik
- Department of Systematic Zoology, Institute of Environmental Biology, Adam Mickiewicz University, 61-712 Poznan, Poland;
| | - Peter Vogel
- Department of Ecology and Evolution, University of Lausanne, 1015 Lausanne, Switzerland;
| | - Ruzanna Djavadian
- Nencki Institute of Experimental Biology Polish Academy of Sciences, 02-093 Warsaw, Poland; (K.B.); (B.T.)
| |
Collapse
|
40
|
Mouihate A, Kalakh S. Maternal Interleukin-6 Hampers Hippocampal Neurogenesis in Adult Rat Offspring in a Sex-Dependent Manner. Dev Neurosci 2021; 43:106-115. [PMID: 34023825 DOI: 10.1159/000516370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/19/2021] [Indexed: 11/19/2022] Open
Abstract
Maternal immune activation (MIA) during pregnancy leads to long-lasting effects on brain development and function. Several lines of evidence suggest that the maternal inflammatory cytokine interleukin (IL)-6 plays a crucial role in the long-lasting effects of MIA on adult offspring. IL-6 is naturally produced during pregnancy in the absence of any underlying immune activation. The objective of this study was to assess whether this naturally occurring IL-6 has long-lasting effects on brain plasticity and function. Therefore, pregnant rats were given either an IL-6-neutralizing antibody (IL-6Ab) or vehicle during the third week of pregnancy. Newly born (doublecortin) and mature neurons (NeuN) were monitored in the hippocampus of adult male and female offspring. Prenatal IL-6Ab led to an enhanced number of newly born and mature neurons in the dentate gyrus of the hippocampus of male but not female adult offspring. This enhanced neurogenesis was associated with an increased propensity in memory acquisition in male offspring. Blunting the naturally occurring IL-6 during pregnancy did not have a significant long-lasting impact on astrocyte cell density (GFAP), or on anxiety-like behavior as assessed with elevated plus maze and open field tests. Taken together, these data suggest that maternal IL-6 contributes, at least in part, to the programming of the brain's development in a sex-dependent manner.
Collapse
Affiliation(s)
- Abdeslam Mouihate
- Department of Physiology, Faculty of Medicine, Kuwait University, Safat, Kuwait
| | - Samah Kalakh
- Department of Physiology, Faculty of Medicine, Kuwait University, Safat, Kuwait
| |
Collapse
|
41
|
de Souza Cardoso J, Baldissarelli J, Reichert KP, Teixeira FC, Pereira Soares MS, Chitolina Schetinger MR, Morsch VM, Farias Martins Filho AO, Duarte Junior HR, Ribeiro Coriolano FH, Spanevello RM, Stefanello FM, Tavares RG. Neuroprotection elicited by resveratrol in a rat model of hypothyroidism: Possible involvement of cholinergic signaling and redox status. Mol Cell Endocrinol 2021; 524:111157. [PMID: 33421531 DOI: 10.1016/j.mce.2021.111157] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 12/29/2020] [Accepted: 12/30/2020] [Indexed: 12/17/2022]
Abstract
Both the cholinergic pathway and oxidative stress are important mechanisms involved in the pathogenesis of hypothyroidism, a condition characterized by low levels of thyroid hormone that predispose the patient to brain dysfunction. Phenolic compounds have numerous health benefits, including antioxidant activity. This study evaluates the preventive effects of resveratrol in the cholinergic system and redox status in rats with methimazole-induced hypothyroidism. Hypothyroidism increases acetylcholinesterase (AChE) activity and density in the cerebral cortex and hippocampus and decreases the α7 and M1 receptor densities in the hippocampus. Hypothyroidism also increases cellular levels of reactive oxygen species (ROS) and thiobarbituric acid reactive substances (TBARS), but reduces total thiol content, and catalase and superoxide dismutase activities in the serum. In the cerebral cortex and hippocampus, hypothyroidism increases the levels of ROS and nitrites. In this study, resveratrol (50 mg/kg) treatment prevents the observed increase in AChE in the cerebral cortex, and increases the protein levels of NeuN, a marker of mature neurons. Resveratrol also prevents changes in serum ROS levels and brain structure, as well as the levels of TBARS, total thiol content, and serum catalase enzyme activity. These collective findings suggest that resveratrol has a high antioxidant capacity and can restore hypothyroidism-triggered alterations related to neurotransmission. Thus, it is a promising agent for the prevention of brain damage resulting from hypothyroidism.
Collapse
Affiliation(s)
- Juliane de Souza Cardoso
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário Capão do Leão s/n, Pelotas, RS, Brazil
| | - Jucimara Baldissarelli
- Departamento de Fisiologia e Farmacologia, Instituto de Biologia, Universidade Federal de Pelotas, Campus Universitário Capão do Leão, Pelotas, RS, Brazil
| | - Karine Paula Reichert
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Fernanda Cardoso Teixeira
- Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário Capão do Leão, Pelotas, RS, Brazil
| | - Mayara Sandrielly Pereira Soares
- Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário Capão do Leão, Pelotas, RS, Brazil
| | - Maria Rosa Chitolina Schetinger
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | - Vera Maria Morsch
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica Toxicológica, Centro de Ciências Naturais e Exatas, Universidade Federal de Santa Maria, Santa Maria, RS, Brazil
| | | | | | | | - Roselia Maria Spanevello
- Laboratório de Neuroquímica, Inflamação e Câncer, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário Capão do Leão, Pelotas, RS, Brazil
| | - Francieli Moro Stefanello
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário Capão do Leão s/n, Pelotas, RS, Brazil.
| | - Rejane Giacomelli Tavares
- Laboratório de Biomarcadores, Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Universidade Federal de Pelotas, Campus Universitário Capão do Leão s/n, Pelotas, RS, Brazil; CBIOS- Universidade Lusófona de Lisboa, Lisboa, Portugal
| |
Collapse
|
42
|
Huckleberry KA, Shansky RM. The unique plasticity of hippocampal adult-born neurons: Contributing to a heterogeneous dentate. Hippocampus 2021; 31:543-556. [PMID: 33638581 DOI: 10.1002/hipo.23318] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/15/2021] [Accepted: 02/09/2021] [Indexed: 12/14/2022]
Abstract
The dentate gyrus (DG) of the hippocampus is evolutionarily conserved as one of the few sites of adult neurogenesis in mammals. Although there is clear evidence that neurogenesis is necessary for healthy hippocampal function, whether adult-born neurons are simply integrated into existing hippocampal networks to serve a similar purpose to that of developmentally born neurons or whether they represent a discrete cell population with unique functions remains less clear. In this review, we consider evidence for discrete cellular, synaptic, and structural features of adult-born DG neurons, suggesting that neurogenesis contributes to the formation of a heterogeneous DG. We therefore propose that hippocampal neurogenesis creates a specialized neuronal subpopulation that may play a key role in hippocampal functions like episodic memory. We note critical gaps in this extensive body of work, including a general failure to include female animals in relevant research and a need for more precise consideration of intrahippocampal neuroanatomy.
Collapse
Affiliation(s)
- Kylie A Huckleberry
- Behavioral Neuroscience Program, Department of Psychology, Northeastern University, Boston, Massachusetts, USA
| | - Rebecca M Shansky
- Behavioral Neuroscience Program, Department of Psychology, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
43
|
Life-long brain compensatory responses to galactic cosmic radiation exposure. Sci Rep 2021; 11:4292. [PMID: 33619310 PMCID: PMC7900210 DOI: 10.1038/s41598-021-83447-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 02/01/2021] [Indexed: 12/02/2022] Open
Abstract
Galactic cosmic radiation (GCR) composed of high-energy, heavy particles (HZE) poses potentially serious hazards to long-duration crewed missions in deep space beyond earth’s magnetosphere, including planned missions to Mars. Chronic effects of GCR exposure on brain structure and cognitive function are poorly understood, thereby limiting risk reduction and mitigation strategies to protect against sequelae from exposure during and after deep-space travel. Given the selective vulnerability of the hippocampus to neurotoxic insult and the importance of this brain region to learning and memory, we hypothesized that GCR-relevant HZE exposure may induce long-term alterations in adult hippocampal neurogenesis, synaptic plasticity, and hippocampal-dependent learning and memory. To test this hypothesis, we irradiated 3-month-old male and female mice with a single, whole-body dose of 10, 50, or 100 cGy 56Fe ions (600 MeV, 181 keV/μm) at Brookhaven National Laboratory. Our data reveal complex, dynamic, time-dependent effects of HZE exposure on the hippocampus. Two months post exposure, neurogenesis, synaptic plasticity and learning were impaired compared to sham-irradiated, age-matched controls. By six months post-exposure, deficits in spatial learning were absent in irradiated mice, and synaptic potentiation was enhanced. Enhanced performance in spatial learning and facilitation of synaptic plasticity in irradiated mice persisted 12 months post-exposure, concomitant with a dramatic rebound in adult-born neurons. Synaptic plasticity and spatial learning remained enhanced 20 months post-exposure, indicating a life-long influence on plasticity and cognition from a single exposure to HZE in young adulthood. These findings suggest that GCR-exposure can persistently alter brain health and cognitive function during and after long-duration travel in deep space.
Collapse
|
44
|
Houben S, Homa M, Yilmaz Z, Leroy K, Brion JP, Ando K. Tau Pathology and Adult Hippocampal Neurogenesis: What Tau Mouse Models Tell us? Front Neurol 2021; 12:610330. [PMID: 33643196 PMCID: PMC7902892 DOI: 10.3389/fneur.2021.610330] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/06/2021] [Indexed: 12/18/2022] Open
Abstract
Adult hippocampal neurogenesis (AHN) has been widely confirmed in mammalian brains. A growing body of evidence points to the fact that AHN sustains hippocampal-dependent functions such as learning and memory. Impaired AHN has been reported in post-mortem human brain hippocampus of Alzheimer's disease (AD) and is considered to contribute to defects in learning and memory. Neurofibrillary tangles (NFTs) and amyloid plaques are the two key neuropathological hallmarks of AD. NFTs are composed of abnormal tau proteins accumulating in many brain areas during the progression of the disease, including in the hippocampus. The physiological role of tau and impact of tau pathology on AHN is still poorly understood. Modifications in AHN have also been reported in some tau transgenic and tau-deleted mouse models. We present here a brief review of advances in the relationship between development of tau pathology and AHN in AD and what insights have been gained from studies in tau mouse models.
Collapse
Affiliation(s)
- Sarah Houben
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Mégane Homa
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Zehra Yilmaz
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Karelle Leroy
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Pierre Brion
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Kunie Ando
- Laboratory of Histology, Neuroanatomy and Neuropathology, UNI (ULB Neuroscience Institute), Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
45
|
Samodien E, Chellan N. Hypothalamic neurogenesis and its implications for obesity-induced anxiety disorders. Front Neuroendocrinol 2021; 60:100871. [PMID: 32976907 DOI: 10.1016/j.yfrne.2020.100871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/08/2020] [Accepted: 09/13/2020] [Indexed: 01/14/2023]
Abstract
Obesity and anxiety are public health problems that have no effective cure. Obesity-induced anxiety is also the most common behavioural trait exhibited amongst obese patients, with the mechanisms linking these disorders being poorly understood. The hypothalamus and hippocampus are reciprocally connected, important neurogenic brain regions that could be vital to understanding these disorders. Dietary, physical activity and lifestyle interventions have been shown to be able to enhance neurogenesis within the hippocampus, while the effects thereof within the hypothalamus is yet to be ascertained. This review describes hypothalamic neurogenesis, its impairment in obesity as well as the effect of interventional therapies. Obesity is characterized by a neurogenic shift towards neuropeptide Y neurons, promoting appetite and weight gain. While, nutraceuticals and exercise promote proopiomelanocortin neuron proliferation, causing diminished appetite and reduced weight gain. Through the furthered development of multimodal approaches targeting both these brain regions could hold an even greater therapeutic potential.
Collapse
Affiliation(s)
- Ebrahim Samodien
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, Cape Town, South Africa.
| | - Nireshni Chellan
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, Cape Town, South Africa; Department of Medical Physiology, Stellenbosch University, Tygerberg, Cape Town, South Africa
| |
Collapse
|
46
|
Schatz M, Saravanan S, d'Adesky ND, Bramlett H, Perez-Pinzon MA, Raval AP. Osteocalcin, ovarian senescence, and brain health. Front Neuroendocrinol 2020; 59:100861. [PMID: 32781196 DOI: 10.1016/j.yfrne.2020.100861] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/20/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022]
Abstract
Menopause, an inevitable event in a woman's life, significantly increases risk of bone resorption and diseases such as Alzheimer's, vascular dementia, cardiac arrest, and stroke. The sole role of bones, as traditionally regarded, is to provide structural support for skeletal muscles and allow for ambulation, however this concept is becoming quickly outdated. New literature has emerged that suggests the bone cell-derived hormone osteocalcin (OCN) plays a pivotal role in cognition. OCN levels are correlated with bone mass density and bone turnover, and thus are strongly influenced by the changes associated with menopause. The goal of the current review is to discuss potential gaps in our knowledge of OCN and cognition, discrepancies in methods of OCN quantification, and therapies to enhance circulating OCN. A discussion on implementing exercise or low frequency vibration interventions at the menopausal transition to reduce risk and severity of neurological diseases and associated cognitive decline is included.
Collapse
Affiliation(s)
- Marc Schatz
- Department of Orthopaedic Surgery, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Sharnikha Saravanan
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, Miami, FL 33136, USA
| | - Nathan D d'Adesky
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, Miami, FL 33136, USA
| | - Helen Bramlett
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL 33136, USA
| | - Miguel A Perez-Pinzon
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, Miami, FL 33136, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory, Department of Neurology, Miami, FL 33136, USA.
| |
Collapse
|