1
|
Mansour H, Azrak R, Cook JJ, Hornburg KJ, Qi Y, Tian Y, Williams RW, Yeh FC, White LE, Johnson GA. The Duke Mouse Brain Atlas: MRI and light sheet microscopy stereotaxic atlas of the mouse brain. SCIENCE ADVANCES 2025; 11:eadq8089. [PMID: 40305623 PMCID: PMC12042906 DOI: 10.1126/sciadv.adq8089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 03/26/2025] [Indexed: 05/02/2025]
Abstract
Atlases of the brain are critical resources that make it possible to share data in a common reference frame. Unexpectedly, there is no three-dimensional (3D) stereotaxic atlas of the mouse brain that provides whole brain coverage at macro to single-cell levels. Diffusion tensor images from five perfusion-fixed (in skull) specimens were acquired at 15 micrometers, the highest resolution ever reported. Diffusion tensor imaging yields multiple 3D volumes, each of which highlights unique cytoarchitecture. The averages were mapped into micro-computed tomography of the mouse skull to create external landmarks (bregma and lambda). Light sheet images of the same brains were coregistered, providing cell maps in the same stereotaxic space. The Allen Reference Atlas was registered to the volume to correct the geometric distortion in that atlas and bring it into the stereotaxic space. The resulting multiscalar (13 terabytes) atlas provides a common spatial framework to anneal data across molecular, structural, and functional studies of mice.
Collapse
Affiliation(s)
- Harrison Mansour
- Duke Center for In Vivo Microscopy, Departments of Radiology and Biomedical Engineering, Duke University, Durham, NC, USA
| | - Ryan Azrak
- Duke Center for In Vivo Microscopy, Departments of Radiology and Biomedical Engineering, Duke University, Durham, NC, USA
| | - James J. Cook
- Duke Center for In Vivo Microscopy, Departments of Radiology and Biomedical Engineering, Duke University, Durham, NC, USA
| | - Kathryn J. Hornburg
- Duke Center for In Vivo Microscopy, Departments of Radiology and Biomedical Engineering, Duke University, Durham, NC, USA
| | - Yi Qi
- Duke Center for In Vivo Microscopy, Departments of Radiology and Biomedical Engineering, Duke University, Durham, NC, USA
| | - Yuqi Tian
- Duke Center for In Vivo Microscopy, Departments of Radiology and Biomedical Engineering, Duke University, Durham, NC, USA
| | - Robert W. Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Fang-Cheng Yeh
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Leonard E. White
- Duke Center for In Vivo Microscopy, Departments of Radiology and Biomedical Engineering, Duke University, Durham, NC, USA
- Department of Neurology, Duke University, Durham, NC, USA
| | - G. Allan Johnson
- Duke Center for In Vivo Microscopy, Departments of Radiology and Biomedical Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
2
|
Barrett CME, Zeidy Z, Farrell A, Cahill LS, Wadden KP. Maternal brain plasticity, physiology and exercise science: A scoping narrative review. Front Neuroendocrinol 2025; 77:101185. [PMID: 39978421 DOI: 10.1016/j.yfrne.2025.101185] [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: 10/10/2024] [Revised: 02/06/2025] [Accepted: 02/15/2025] [Indexed: 02/22/2025]
Abstract
INTRODUCTION The perinatal period is characterized by extreme shifts in hormones, neurochemistry, and life experiences that drive significant changes in the brain, known as maternal plasticity. Due to rising maternal health conditions, such as postpartum depression, there is a critical need to investigate factors, such as engagement in physical activity and exercise, that may mitigate susceptibility to maladaptive maternal plasticity. This scoping review aims to analyze exercise interventions and maternal brain outcomes during reproduction. METHODS A systematic search was completed in Medline, Embase, CINAHL, PsycINFO, SportDiscuss. The key concepts of the search were (i) brain plasticity, (ii) maternal reproductive period including pre-conception, pregnancy, and postpartum, and (iii) exercise interventions. Due to the limited amount of evidence available on this topic, the review findings were discussed using a combined scoping and narrative review approach. RESULTS The search produced 2,167 unique articles after removing 2588 duplicates. Covidence software was used for the screening procedure. Following title and abstract screening, 2160 articles were deemed irrelevant and removed. Seven articles moved forward to full-text screening. One article was excluded during full-text screening for wrong outcomes, leaving six papers for extraction. Extraction revealed that four out of six studies were conducted in the rodent alone, one was conducted in humans alone and one was conducted in both a human and a rodent model. DISCUSSION The methodological inconsistencies in the limited number of studies within this field highlight the need for standardization, which motivated the development of the Consensus on Exercise Reporting Template for animal research. Moreover, the present review highlights future directions and knowledge gaps, emphasizing the critical need for high-quality research to address the many unanswered questions regarding the impact of exercise on the maternal brain.
Collapse
Affiliation(s)
- Catherine M E Barrett
- School of Human Kinetics and Recreation, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Zohreh Zeidy
- School of Human Kinetics and Recreation, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Alison Farrell
- Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Lindsay S Cahill
- Department of Chemistry, Faculty of Science, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Katie P Wadden
- School of Human Kinetics and Recreation, Memorial University of Newfoundland, St. John's, NL, Canada.
| |
Collapse
|
3
|
Wang SS, Peng Y, Fan PL, Ye JR, Ma WY, Wu QL, Wang HY, Tian YJ, He WB, Yan X, Zhang Z, Chu SF, Chen NH. Ginsenoside Rg1 ameliorates stress-exacerbated Parkinson's disease in mice by eliminating RTP801 and α-synuclein autophagic degradation obstacle. Acta Pharmacol Sin 2025; 46:308-325. [PMID: 39227736 PMCID: PMC11747340 DOI: 10.1038/s41401-024-01374-w] [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: 04/29/2024] [Accepted: 07/31/2024] [Indexed: 09/05/2024]
Abstract
Emerging evidence shows that psychological stress promotes the progression of Parkinson's disease (PD) and the onset of dyskinesia in non-PD individuals, highlighting a potential avenue for therapeutic intervention. We previously reported that chronic restraint-induced psychological stress precipitated the onset of parkinsonism in 10-month-old transgenic mice expressing mutant human α-synuclein (αSyn) (hαSyn A53T). We refer to these as chronic stress-genetic susceptibility (CSGS) PD model mice. In this study we investigated whether ginsenoside Rg1, a principal compound in ginseng notable for soothing the mind, could alleviate PD deterioration induced by psychological stress. Ten-month-old transgenic hαSyn A53T mice were subjected to 4 weeks' restraint stress to simulate chronic stress conditions that worsen PD, meanwhile the mice were treated with Rg1 (40 mg· kg-1 ·d-1, i.g.), and followed by functional magnetic resonance imaging (fMRI) and a variety of neurobehavioral tests. We showed that treatment with Rg1 significantly alleviated both motor and non-motor symptoms associated with PD. Functional MRI revealed that Rg1 treatment enhanced connectivity between brain regions implicated in PD, and in vivo multi-channel electrophysiological assay showed improvements in dyskinesia-related electrical activity. In addition, Rg1 treatment significantly attenuated the degeneration of dopaminergic neurons and reduced the pathological aggregation of αSyn in the striatum and SNc. We revealed that Rg1 treatment selectively reduced the level of the stress-sensitive protein RTP801 in SNc under chronic stress conditions, without impacting the acute stress response. HPLC-MS/MS analysis coupled with site-directed mutation showed that Rg1 promoted the ubiquitination and subsequent degradation of RTP801 at residues K188 and K218, a process mediated by the Parkin RING2 domain. Utilizing αSyn A53T+; RTP801-/- mice, we confirmed the critical role of RTP801 in stress-aggravated PD and its necessity for Rg1's protective effects. Moreover, Rg1 alleviated obstacles in αSyn autophagic degradation by ameliorating the RTP801-TXNIP-mediated deficiency of ATP13A2. Collectively, our results suggest that ginsenoside Rg1 holds promise as a therapeutic choice for treating PD-sensitive individuals who especially experience high levels of stress and self-imposed expectations.
Collapse
Affiliation(s)
- Sha-Sha Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Ye Peng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- School of Pharmacy, Minzu University of China, Beijing, 100081, China
| | - Ping-Long Fan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Jun-Rui Ye
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Wen-Yu Ma
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Qing-Lin Wu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Hong-Yun Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Ya-Juan Tian
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030024, China
| | - Wen-Bin He
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, National International Joint Research Center for Molecular Chinese Medicine, Shanxi University of Chinese Medicine, Taiyuan, 030024, China
| | - Xu Yan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Shi-Feng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
4
|
Qi J, Suo X, Tian C, Xia X, Qin W, Wang P, Tang J, Xu J, Fu J, Liu N, Yu C, Shen H, Dou Y. TESC overexpression mitigates amyloid-β-induced hippocampal atrophy and memory decline. Gene 2025; 933:148939. [PMID: 39278373 DOI: 10.1016/j.gene.2024.148939] [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: 05/16/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND AND OBJECTIVES Genome-wide association studies (GWASs) have identified numerous candidate genes for human brain-imaging phenotypes; however, the biological relevance of many of these genes remains unconfirmed. This study aimed to investigate the causal relationships among tescalcin (TESC) (a GWAS-indicated gene), hippocampal volume, Alzheimer's disease (AD), and the underlying biological mechanisms. METHODS Human transcriptional data were analyzed to confirm relative TESC expression in the hippocampus. In cell experiments, RNA-seq analysis was used to identify the potential biological pathways for TESC overexpression, and immunofluorescence imaging and cell viability assays were used to evaluate the effect of TESC overexpression on neuronal structure and survival. In animal experiments, the effects of TESC overexpression on hippocampal volume and cognitive function in normal mice and amyloid-β (Aβ)-induced AD mice were investigated by 9.4 T magnetic resonance imaging and behavioral tests. Underlying mechanisms were further assessed via western blotting and electrophysiological recordings. RESULTS Human transcriptional data demonstrated that TESC is primarily expressed in the hippocampus and neurons. TESC overexpression enhanced the viability of HT22 cells and reduced Aβ-induced cell death. In mouse models, Tesc-overexpressing mice revealed increased hippocampal volume, likely owing to enhanced cell viability and long-term potentiation (LTP), and reducing apoptotic- and oxidation-induced hippocampal damage. TESC overexpression could significantly mitigate Aβ-induced hippocampal atrophy and memory impairment, potentially by reducing Aβ-induced neuronal apoptosis and LTP weakening. CONCLUSION This study exemplifies the translation of GWAS findings into actionable biological knowledge and suggests that upregulation of TESC may offer a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Jinbo Qi
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Xinjun Suo
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, PR China; School of Medical Technology, Tianjin Medical University, Tianjin 300070, PR China
| | - Chunxiao Tian
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin 300070, PR China
| | - Xianyou Xia
- Department of Cell Biology, School of Basic Medicine and Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin 300070, PR China
| | - Wen Qin
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Ping Wang
- School of Medical Technology, Tianjin Medical University, Tianjin 300070, PR China
| | - Jie Tang
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Jiayuan Xu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Jilian Fu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Nana Liu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Chunshui Yu
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, PR China; School of Medical Technology, Tianjin Medical University, Tianjin 300070, PR China
| | - Hui Shen
- Department of Cell Biology, School of Basic Medicine and Tianjin Key Laboratory of Medical Epigenetics, Tianjin Medical University, Tianjin 300070, PR China.
| | - Yan Dou
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging & Tianjin Institute of Radiology, Tianjin Medical University General Hospital, Tianjin 300052, PR China.
| |
Collapse
|
5
|
Pritschet L, Taylor CM, Cossio D, Faskowitz J, Santander T, Handwerker DA, Grotzinger H, Layher E, Chrastil ER, Jacobs EG. Neuroanatomical changes observed over the course of a human pregnancy. Nat Neurosci 2024; 27:2253-2260. [PMID: 39284962 PMCID: PMC11537970 DOI: 10.1038/s41593-024-01741-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 07/29/2024] [Indexed: 09/25/2024]
Abstract
Pregnancy is a period of profound hormonal and physiological changes experienced by millions of women annually, yet the neural changes unfolding in the maternal brain throughout gestation are not well studied in humans. Leveraging precision imaging, we mapped neuroanatomical changes in an individual from preconception through 2 years postpartum. Pronounced decreases in gray matter volume and cortical thickness were evident across the brain, standing in contrast to increases in white matter microstructural integrity, ventricle volume and cerebrospinal fluid, with few regions untouched by the transition to motherhood. This dataset serves as a comprehensive map of the human brain across gestation, providing an open-access resource for the brain imaging community to further explore and understand the maternal brain.
Collapse
Affiliation(s)
- Laura Pritschet
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA, USA.
| | - Caitlin M Taylor
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA, USA
| | - Daniela Cossio
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Joshua Faskowitz
- Section on Functional Imaging Methods, Laboratory of Brain and Cognition, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Tyler Santander
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA, USA
| | - Daniel A Handwerker
- Section on Functional Imaging Methods, Laboratory of Brain and Cognition, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Hannah Grotzinger
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA, USA
| | - Evan Layher
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA, USA
| | - Elizabeth R Chrastil
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA.
| | - Emily G Jacobs
- Department of Psychological & Brain Sciences, University of California, Santa Barbara, CA, USA.
- Neuroscience Research Institute, University of California, Santa Barbara, CA, USA.
| |
Collapse
|
6
|
Magalhães R, Marques F, Selingue E, Boumezbeur F, Mériaux S, Sousa N. A longitudinal MRI analysis reveals altered brain connectivity and microstructural changes in a transgenic mouse model of Alzheimer's disease. Neurobiol Dis 2024; 201:106679. [PMID: 39321859 DOI: 10.1016/j.nbd.2024.106679] [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: 07/29/2024] [Revised: 09/17/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive cognitive decline and neuropathological changes, yet the underlying neurobiological mechanisms remain elusive. Here, we employed a multimodal longitudinal neuroimaging approach, using anatomical and functional sequences on a high field magnetic resonance imaging (MRI) preclinical scanner, to investigate alterations in brain connectivity and white matter microstructure in a transgenic mouse model of AD (J20) when compared to wild-type (WT) littermates. Functional connectivity analysis revealed distinct network disruptions in J20 mice, primarily involving connections between posterior and anterior brain regions; importantly, a significant interaction between group and age highlighted an exacerbation of these connectivity changes with advancing age in J20 mice. In addition, significant reductions in fractional anisotropy (FA) were observed in the corpus callosum of J20 mice compared to WT, indicative of microstructural alterations consistent with white matter pathology. The observed alterations in brain connectivity and microstructure provide valuable insights into the spatiotemporal processes underlying AD-related decline and underscore the utility of multimodal neuroimaging in elucidating the neurobiological substrates of AD pathology in animal models.
Collapse
Affiliation(s)
- Ricardo Magalhães
- NeuroSpin, Paris-Saclay University, CEA, CNRS, 91191 Gif-sur-Yvette, France; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.; ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Fernanda Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.; ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Erwan Selingue
- NeuroSpin, Paris-Saclay University, CEA, CNRS, 91191 Gif-sur-Yvette, France
| | - Fawzi Boumezbeur
- NeuroSpin, Paris-Saclay University, CEA, CNRS, 91191 Gif-sur-Yvette, France
| | - Sébastien Mériaux
- NeuroSpin, Paris-Saclay University, CEA, CNRS, 91191 Gif-sur-Yvette, France
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.; ICVS/3B's, PT Government Associate Laboratory, Braga, Guimarães, Portugal; Clinical Academic Center Braga (2CA-Braga), Braga, Portugal.
| |
Collapse
|
7
|
Dufford AJ, Patterson G, Kim P. Longitudinal neuroanatomical increases from early to one-year postpartum. Brain Struct Funct 2024:10.1007/s00429-024-02852-x. [PMID: 39299954 DOI: 10.1007/s00429-024-02852-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/13/2024] [Indexed: 09/22/2024]
Abstract
Preclinical studies have provided causal evidence that the postpartum period involves regional neuroanatomical changes in 'maternal' brain regions to support the transition to offspring caregiving. Few studies, in humans, have examined neuroanatomical changes from early to one-year postpartum with longitudinal neuroimaging data and their association with postpartum mood changes. In the present study, we examined longitudinal changes in surface morphometry (cortical thickness and surface area) in regions previously implicated in the transition to parenthood. We also examined longitudinal volumetric neuroanatomical changes in three subcortical regions of the maternal brain: the hippocampus, amygdala, and ventral diencephalon. Twenty-four participants underwent longitudinal structural magnetic resonance imaging at 1-4 weeks and 1 year postpartum. Cortical thickness increased from early to one-year postpartum in the left (p = .003, Bonferroni corrected) and right (p = .02, Bonferroni corrected) superior frontal gyrus. No significant increases (or decreases) were observed in these regions for surface area. Volumetric increases, across the postpartum period, were found in the left amygdala (p = .001, Bonferroni corrected) and right ventral diencephalon (p = .01, Bonferroni corrected). An exploratory analysis of depressive symptoms found reductions in depressive symptoms from early postpartum to one-year postpartum were associated with greater cortical thickness in the superior frontal gyrus for both the left (p = .02) and right (p = .02) hemispheres. The findings expand our evidence of the neuroanatomical changes that occur across the postpartum period in humans and motivate future studies to examine how mood changes across this period are associated with cortical thickness of the superior frontal gyrus.
Collapse
Affiliation(s)
- Alexander J Dufford
- Center for Mental Health Innovation, Oregon Health & Science University, Portland, OR, USA
- Department of Psychiatry, Oregon Health & Science University, Portland, OR, USA
| | | | - Pilyoung Kim
- Department of Psychology, University of Denver, Denver, CO, 80210, USA.
- Department of Psychology, Ewha Womans University, Seoul, South Korea.
| |
Collapse
|
8
|
de Lange AMG, Leonardsen EH, Barth C, Schindler LS, Crestol A, Holm MC, Subramaniapillai S, Hill D, Alnæs D, Westlye LT. Parental status and markers of brain and cellular age: A 3D convolutional network and classification study. Psychoneuroendocrinology 2024; 165:107040. [PMID: 38636355 DOI: 10.1016/j.psyneuen.2024.107040] [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: 01/20/2024] [Revised: 03/29/2024] [Accepted: 03/30/2024] [Indexed: 04/20/2024]
Abstract
Recent research shows prominent effects of pregnancy and the parenthood transition on structural brain characteristics in humans. Here, we present a comprehensive study of how parental status and number of children born/fathered links to markers of brain and cellular ageing in 36,323 UK Biobank participants (age range 44.57-82.06 years; 52% female). To assess global effects of parenting on the brain, we trained a 3D convolutional neural network on T1-weighted magnetic resonance images, and estimated brain age in a held-out test set. To investigate regional specificity, we extracted cortical and subcortical volumes using FreeSurfer, and ran hierarchical clustering to group regional volumes based on covariance. Leukocyte telomere length (LTL) derived from DNA was used as a marker of cellular ageing. We employed linear regression models to assess relationships between number of children, brain age, regional brain volumes, and LTL, and included interaction terms to probe sex differences in associations. Lastly, we used the brain measures and LTL as features in binary classification models, to determine if markers of brain and cellular ageing could predict parental status. The results showed associations between a greater number of children born/fathered and younger brain age in both females and males, with stronger effects observed in females. Volume-based analyses showed maternal effects in striatal and limbic regions, which were not evident in fathers. We found no evidence for associations between number of children and LTL. Classification of parental status showed an Area under the ROC Curve (AUC) of 0.57 for the brain age model, while the models using regional brain volumes and LTL as predictors showed AUCs of 0.52. Our findings align with previous population-based studies of middle- and older-aged parents, revealing subtle but significant associations between parental experience and neuroimaging-based surrogate markers of brain health. The findings further corroborate results from longitudinal cohort studies following parents across pregnancy and postpartum, potentially indicating that the parenthood transition is associated with long-term influences on brain health.
Collapse
Affiliation(s)
- Ann-Marie G de Lange
- Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland; Department of Psychology, University of Oslo, Oslo, Norway; Department of Psychiatry, University of Oxford, Oxford, UK.
| | | | - Claudia Barth
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Louise S Schindler
- Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland; Department of Psychology, University of Oslo, Oslo, Norway; Department of Psychiatry, University of Oxford, Oxford, UK
| | - Arielle Crestol
- Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | | | - Sivaniya Subramaniapillai
- Department of Clinical Neurosciences, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland; Department of Psychology, University of Oslo, Oslo, Norway
| | - Dónal Hill
- Swiss Data Science Center (SDSC), EPFL-ETHZ, Switzerland
| | - Dag Alnæs
- Department of Psychology, University of Oslo, Oslo, Norway; Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lars T Westlye
- Department of Psychology, University of Oslo, Oslo, Norway; Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo, Oslo, Norway
| |
Collapse
|
9
|
Dufford A, Patterson G, Kim P. Longitudinal Neuroanatomical Increases from Early to One-Year Postpartum. RESEARCH SQUARE 2024:rs.3.rs-4432804. [PMID: 38883787 PMCID: PMC11178002 DOI: 10.21203/rs.3.rs-4432804/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Preclinical studies have provided causal evidence that the postpartum period involves regional neuroanatomical changes in 'maternal' brain regions to support the transition to offspring caregiving. Few studies, in humans, have examined neuroanatomical changes from early to one-year postpartum with longitudinal neuroimaging data and their association with postpartum mood changes. In this study, we examined longitudinal changes in surface morphometry (cortical thickness and surface area) in regions previously implicated in the transition to parenthood. We also examined longitudinal volumetric neuroanatomical changes in three subcortical regions of the maternal brain: the hippocampus, amygdala, and ventral diencephalon. Twenty-four participants underwent longitudinal structural magnetic resonance imaging at 2-4 weeks and 1 year postpartum. Cortical thickness increased from early to one-year postpartum in the left (p = .003, Bonferroni corrected) and right (p = .02, Bonferroni corrected) superior frontal gyrus. No significant increases (or decreases) were observed in these regions for surface area. Volumetric increases, across the postpartum period, were found in the left amygdala (p = .001, Bonferroni corrected) and right ventral diencephalon (p = .01, Bonferroni corrected). An exploratory analysis of depressive symptoms found reductions in depressive symptoms from early postpartum to one-year postpartum were associated with greater cortical thickness in the superior frontal gyrus for both the left (p = .02) and right (p = .02) hemispheres. The findings expand our evidence of the neuroanatomical changes that occur across the postpartum period in humans and motivate future studies to examine how mood changes across this period are associated with cortical thickness of the superior frontal gyrus.
Collapse
|
10
|
Spalek K, Straathof M, Koyuncu L, Grydeland H, van der Geest A, Van't Hof SR, Crone EA, Barba-Müller E, Carmona S, Denys D, Tamnes CK, Burke S, Hoekzema E. Pregnancy renders anatomical changes in hypothalamic substructures of the human brain that relate to aspects of maternal behavior. Psychoneuroendocrinology 2024; 164:107021. [PMID: 38492349 DOI: 10.1016/j.psyneuen.2024.107021] [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: 08/04/2023] [Revised: 03/07/2024] [Accepted: 03/09/2024] [Indexed: 03/18/2024]
Abstract
Animal studies have shown that pregnancy is associated with neural adaptations that promote maternal care. The hypothalamus represents a central structure of the mammalian maternal brain and hormonal priming of specific hypothalamic nuclei plays a key role in the induction and expression of maternal behavior. In humans, we have previously demonstrated that becoming a mother involves changes in grey matter anatomy, primarily in association areas of the cerebral cortex. In the current study, we investigated whether pregnancy renders anatomical changes in the hypothalamus. Using an advanced delineation technique, five hypothalamic substructures were defined in longitudinal MRI scans of 107 women extracted from two prospective pre-conception cohort studies, including 50 women who were scanned before and after pregnancy and 57 nulliparous control women scanned at a similar time interval. We showed that becoming a mother is associated with volume reductions in the anterior-superior, superior tuberal and posterior hypothalamus. In addition, these structural changes related to hormonal levels during pregnancy and specific aspects of self-reported maternal behavior in late pregnancy, including maternal-fetal attachment and nesting behavior. These findings show that pregnancy leads to changes in hypothalamic anatomy and suggest that these contribute to the development of maternal behavior in humans, supporting the conservation of key aspects of maternal brain circuitry and their role in maternal behavior across species.
Collapse
Affiliation(s)
- Klara Spalek
- Hoekzema Lab, Amsterdam University Medical Center (Amsterdam UMC), location University of Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam Reproduction and Development, Amsterdam, the Netherlands
| | - Milou Straathof
- Hoekzema Lab, Amsterdam University Medical Center (Amsterdam UMC), location University of Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam Reproduction and Development, Amsterdam, the Netherlands
| | - Lal Koyuncu
- Brain and Development Research Center, Leiden Institute for Brain and Cognition, Leiden University, Leiden, the Netherlands
| | - Håkon Grydeland
- Center for Lifespan Changes in Brain and Cognition (LCBC), Department of Psychology, University of Oslo, Norway
| | - Anouk van der Geest
- Brain and Development Research Center, Leiden Institute for Brain and Cognition, Leiden University, Leiden, the Netherlands
| | - Sophie R Van't Hof
- Hoekzema Lab, Amsterdam University Medical Center (Amsterdam UMC), location University of Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam Reproduction and Development, Amsterdam, the Netherlands
| | - Eveline A Crone
- Brain and Development Research Center, Leiden Institute for Brain and Cognition, Leiden University, Leiden, the Netherlands
| | | | - Susana Carmona
- Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Damiaan Denys
- Department of Psychiatry, Amsterdam University Medical Center (Amsterdam UMC), location University of Amsterdam, the Netherlands
| | - Christian K Tamnes
- PROMENTA Research Center, Department of Psychology, University of Oslo, Oslo, Norway; NORMENT, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Psychiatric Research, Diakonhjemmet Hospital, Oslo, Norway
| | - Sarah Burke
- Brain and Development Research Center, Leiden Institute for Brain and Cognition, Leiden University, Leiden, the Netherlands; Interdisciplinary Center Psychopathology and Emotion regulation, Department of Psychiatry, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Elseline Hoekzema
- Hoekzema Lab, Amsterdam University Medical Center (Amsterdam UMC), location University of Amsterdam, Department of Psychiatry, Amsterdam Neuroscience, Amsterdam Reproduction and Development, Amsterdam, the Netherlands.
| |
Collapse
|
11
|
Pritschet L, Taylor CM, Cossio D, Santander T, Grotzinger H, Faskowitz J, Handwerker DA, Layher E, Chrastil ER, Jacobs EG. Neuroanatomical changes observed over the course of a human pregnancy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.14.571688. [PMID: 38168195 PMCID: PMC10760186 DOI: 10.1101/2023.12.14.571688] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Pregnancy is a period of profound hormonal and physiological change experienced by millions of women annually, yet the neural changes unfolding in the maternal brain throughout gestation have not been studied in humans. Leveraging precision imaging, we mapped neuroanatomical changes in an individual from preconception through two years postpartum. Pronounced decreases in gray matter volume and cortical thickness were evident across the brain, which stand in contrast to increases in white matter microstructural integrity, ventricle volume, and cerebrospinal fluid, with few regions untouched by the transition to motherhood. This dataset serves as the first comprehensive map of the human brain across gestation, providing an open-access resource for the brain imaging community to stimulate further exploration and discovery.
Collapse
|
12
|
Ma X, Xing Y, Zhai R, Du Y, Yan H. Development and advancements in rodent MRI-based brain atlases. Heliyon 2024; 10:e27421. [PMID: 38510053 PMCID: PMC10950579 DOI: 10.1016/j.heliyon.2024.e27421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/15/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
Rodents, particularly mice and rats, are extensively utilized in fundamental neuroscience research. Brain atlases have played a pivotal role in this field, evolving from traditional printed histology atlases to digital atlases incorporating diverse imaging datasets. Magnetic resonance imaging (MRI)-based brain atlases, also known as brain maps, have been employed in specific studies. However, the existence of numerous versions of MRI-based brain atlases has impeded their standardized application and widespread use, despite the consensus within the academic community regarding their significance in mice and rats. Furthermore, there is a dearth of comprehensive and systematic reviews on MRI-based brain atlases for rodents. This review aims to bridge this gap by providing a comprehensive overview of the advancements in MRI-based brain atlases for rodents, with a specific focus on mice and rats. It seeks to explore the advantages and disadvantages of histologically printed brain atlases in comparison to MRI brain atlases, delineate the standardized methods for creating MRI brain atlases, and summarize their primary applications in neuroscience research. Additionally, this review aims to assist researchers in selecting appropriate versions of MRI brain atlases for their studies or refining existing MRI brain atlas resources, thereby facilitating the development and widespread adoption of standardized MRI-based brain atlases in rodents.
Collapse
Affiliation(s)
- Xiaoyi Ma
- Department of Geriatrics, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yao Xing
- School of Information Science and Technology, Fudan University, Shanghai, 200433, China
- Wuhan United Imaging Life Science Instrument Co., Ltd., Wuhan, 430071, China
| | - Renkuan Zhai
- Wuhan United Imaging Life Science Instrument Co., Ltd., Wuhan, 430071, China
| | - Yingying Du
- Wuhan United Imaging Life Science Instrument Co., Ltd., Wuhan, 430071, China
| | - Huanhuan Yan
- Shenzhen United Imaging Research Institute of Innovative Medical Equipment, Shenzhen, 518048, China
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| |
Collapse
|
13
|
Willekens SMA, Morini F, Mediavilla T, Nilsson E, Orädd G, Hahn M, Chotiwan N, Visa M, Berggren PO, Ilegems E, Överby AK, Ahlgren U, Marcellino D. An MR-based brain template and atlas for optical projection tomography and light sheet fluorescence microscopy in neuroscience. Front Neurosci 2024; 18:1328815. [PMID: 38601090 PMCID: PMC11004350 DOI: 10.3389/fnins.2024.1328815] [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: 10/27/2023] [Accepted: 03/11/2024] [Indexed: 04/12/2024] Open
Abstract
Introduction Optical Projection Tomography (OPT) and light sheet fluorescence microscopy (LSFM) are high resolution optical imaging techniques, ideally suited for ex vivo 3D whole mouse brain imaging. Although they exhibit high specificity for their targets, the anatomical detail provided by tissue autofluorescence remains limited. Methods T1-weighted images were acquired from 19 BABB or DBE cleared brains to create an MR template using serial longitudinal registration. Afterwards, fluorescent OPT and LSFM images were coregistered/normalized to the MR template to create fusion images. Results Volumetric calculations revealed a significant difference between BABB and DBE cleared brains, leading to develop two optimized templates, with associated tissue priors and brain atlas, for BABB (OCUM) and DBE (iOCUM). By creating fusion images, we identified virus infected brain regions, mapped dopamine transporter and translocator protein expression, and traced innervation from the eye along the optic tract to the thalamus and superior colliculus using cholera toxin B. Fusion images allowed for precise anatomical identification of fluorescent signal in the detailed anatomical context provided by MR. Discussion The possibility to anatomically map fluorescent signals on magnetic resonance (MR) images, widely used in clinical and preclinical neuroscience, would greatly benefit applications of optical imaging of mouse brain. These specific MR templates for cleared brains enable a broad range of neuroscientific applications integrating 3D optical brain imaging.
Collapse
Affiliation(s)
- Stefanie M. A. Willekens
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Federico Morini
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Tomas Mediavilla
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Emma Nilsson
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Greger Orädd
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Max Hahn
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Nunya Chotiwan
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Montse Visa
- The Rolf Luft Research Centre for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Per-Olof Berggren
- The Rolf Luft Research Centre for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Erwin Ilegems
- The Rolf Luft Research Centre for Diabetes and Endocrinology, Karolinska Institutet, Stockholm, Sweden
| | - Anna K. Överby
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
| | - Ulf Ahlgren
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| | - Daniel Marcellino
- Department of Medical and Translational Biology, Umeå University, Umeå, Sweden
| |
Collapse
|
14
|
Howland MA. Recalibration of the stress response system over adult development: Is there a perinatal recalibration period? Dev Psychopathol 2023; 35:2315-2337. [PMID: 37641984 PMCID: PMC10901284 DOI: 10.1017/s0954579423000998] [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] [Indexed: 08/31/2023]
Abstract
During early life-sensitive periods (i.e., fetal, infancy), the developing stress response system adaptively calibrates to match environmental conditions, whether harsh or supportive. Recent evidence suggests that puberty is another window when the stress system is open to recalibration if environmental conditions have shifted significantly. Whether additional periods of recalibration exist in adulthood remains to be established. The present paper draws parallels between childhood (re)calibration periods and the perinatal period to hypothesize that this phase may be an additional window of stress recalibration in adult life. Specifically, the perinatal period (defined here to include pregnancy, lactation, and early parenthood) is also a developmental switch point characterized by heightened neural plasticity and marked changes in stress system function. After discussing these similarities, lines of empirical evidence needed to substantiate the perinatal stress recalibration hypothesis are proposed, and existing research support is reviewed. Complexities and challenges related to delineating the boundaries of perinatal stress recalibration and empirically testing this hypothesis are discussed, as well as possibilities for future multidisciplinary research. In the theme of this special issue, perinatal stress recalibration may be a mechanism of multilevel, multisystem risk, and resilience, both intra-individually and intergenerationally, with implications for optimizing interventions.
Collapse
Affiliation(s)
- Mariann A Howland
- Institute of Child Development, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
15
|
Du X, Yan Y, Yu J, Zhu T, Huang CC, Zhang L, Shan X, Li R, Dai Y, Lv H, Zhang XY, Feng J, Li WG, Luo Q, Li F. SH2B1 Tunes Hippocampal ERK Signaling to Influence Fluid Intelligence in Humans and Mice. RESEARCH (WASHINGTON, D.C.) 2023; 6:0269. [PMID: 38434247 PMCID: PMC10907025 DOI: 10.34133/research.0269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 10/19/2023] [Indexed: 03/05/2024]
Abstract
Fluid intelligence is a cognitive domain that encompasses general reasoning, pattern recognition, and problem-solving abilities independent of task-specific experience. Understanding its genetic and neural underpinnings is critical yet challenging for predicting human development, lifelong health, and well-being. One approach to address this challenge is to map the network of correlations between intelligence and other constructs. In the current study, we performed a genome-wide association study using fluid intelligence quotient scores from the UK Biobank to explore the genetic architecture of the associations between obesity risk and fluid intelligence. Our results revealed novel common genetic loci (SH2B1, TUFM, ATP2A1, and FOXO3) underlying the association between fluid intelligence and body metabolism. Surprisingly, we demonstrated that SH2B1 variation influenced fluid intelligence independently of its effects on metabolism but partially mediated its association with bilateral hippocampal volume. Consistently, selective genetic ablation of Sh2b1 in the mouse hippocampus, particularly in inhibitory neurons, but not in excitatory neurons, significantly impaired working memory, short-term novel object recognition memory, and behavioral flexibility, but not spatial learning and memory, mirroring the human intellectual performance. Single-cell genetic profiling of Sh2B1-regulated molecular pathways revealed that Sh2b1 deletion resulted in aberrantly enhanced extracellular signal-regulated kinase (ERK) signaling, whereas pharmacological inhibition of ERK signaling reversed the associated behavioral impairment. Our cross-species study thus provides unprecedented insight into the role of SH2B1 in fluid intelligence and has implications for understanding the genetic and neural underpinnings of lifelong mental health and well-being.
Collapse
Affiliation(s)
- Xiujuan Du
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education-Shanghai Key Laboratory for Children’s Environmental Health,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Developmental and Behavioral Pediatric Department,
Shanghai Xinhua Children’s Hospital, Shanghai 200092, China
- National Clinical Research Center for Aging and Medicine at Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, Ministry of Education-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence,
Fudan University, Shanghai 200433, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science and Human Phenom Institute,
Fudan University, Shanghai 200032, China
| | - Yuhua Yan
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education-Shanghai Key Laboratory for Children’s Environmental Health,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Developmental and Behavioral Pediatric Department,
Shanghai Xinhua Children’s Hospital, Shanghai 200092, China
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education),
School of Life Sciences, East China Normal University, Shanghai 200062, China
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science,
Fudan University, Shanghai 200032, China
| | - Juehua Yu
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education-Shanghai Key Laboratory for Children’s Environmental Health,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Developmental and Behavioral Pediatric Department,
Shanghai Xinhua Children’s Hospital, Shanghai 200092, China
- Center for Experimental Studies and Research,
The First Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Tailin Zhu
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education-Shanghai Key Laboratory for Children’s Environmental Health,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Developmental and Behavioral Pediatric Department,
Shanghai Xinhua Children’s Hospital, Shanghai 200092, China
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education),
School of Life Sciences, East China Normal University, Shanghai 200062, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai 201210, China
| | - Chu-Chung Huang
- Institute of Cognitive Neuroscience, School of Psychology and Cognitive Science,
East China Normal University, Shanghai 200062, China
| | - Lingli Zhang
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education-Shanghai Key Laboratory for Children’s Environmental Health,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Developmental and Behavioral Pediatric Department,
Shanghai Xinhua Children’s Hospital, Shanghai 200092, China
| | - Xingyue Shan
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education-Shanghai Key Laboratory for Children’s Environmental Health,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Developmental and Behavioral Pediatric Department,
Shanghai Xinhua Children’s Hospital, Shanghai 200092, China
- Shanghai Key Laboratory of Brain Functional Genomics (Ministry of Education),
School of Life Sciences, East China Normal University, Shanghai 200062, China
| | - Ren Li
- National Clinical Research Center for Aging and Medicine at Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, Ministry of Education-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence,
Fudan University, Shanghai 200433, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science and Human Phenom Institute,
Fudan University, Shanghai 200032, China
| | - Yuan Dai
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education-Shanghai Key Laboratory for Children’s Environmental Health,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Developmental and Behavioral Pediatric Department,
Shanghai Xinhua Children’s Hospital, Shanghai 200092, China
| | - Hui Lv
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education-Shanghai Key Laboratory for Children’s Environmental Health,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Developmental and Behavioral Pediatric Department,
Shanghai Xinhua Children’s Hospital, Shanghai 200092, China
| | - Xiao-Yong Zhang
- National Clinical Research Center for Aging and Medicine at Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, Ministry of Education-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence,
Fudan University, Shanghai 200433, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science and Human Phenom Institute,
Fudan University, Shanghai 200032, China
| | - Jianfeng Feng
- National Clinical Research Center for Aging and Medicine at Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, Ministry of Education-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence,
Fudan University, Shanghai 200433, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science and Human Phenom Institute,
Fudan University, Shanghai 200032, China
| | - Wei-Guang Li
- Department of Rehabilitation Medicine, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science,
Fudan University, Shanghai 200032, China
| | - Qiang Luo
- National Clinical Research Center for Aging and Medicine at Huashan Hospital, Institute of Science and Technology for Brain-Inspired Intelligence, Ministry of Education-Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence,
Fudan University, Shanghai 200433, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Institutes of Brain Science and Human Phenom Institute,
Fudan University, Shanghai 200032, China
| | - Fei Li
- Developmental and Behavioral Pediatric Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and Ministry of Education-Shanghai Key Laboratory for Children’s Environmental Health,
Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Developmental and Behavioral Pediatric Department,
Shanghai Xinhua Children’s Hospital, Shanghai 200092, China
- Shanghai Research Center for Brain Science and Brain-Inspired Intelligence, Shanghai 201210, China
| |
Collapse
|
16
|
Servin-Barthet C, Martínez-García M, Pretus C, Paternina-Die M, Soler A, Khymenets O, Pozo ÓJ, Leuner B, Vilarroya O, Carmona S. The transition to motherhood: linking hormones, brain and behaviour. Nat Rev Neurosci 2023; 24:605-619. [PMID: 37612425 DOI: 10.1038/s41583-023-00733-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2023] [Indexed: 08/25/2023]
Abstract
We are witnessing a stark increase in scientific interest in the neurobiological processes associated with pregnancy and maternity. Convergent evidence suggests that around the time of labour, first-time mothers experience a specific pattern of neuroanatomical changes that are associated with maternal behaviour. Here we provide an overview of the human neurobiological adaptations of motherhood, focusing on the interplay between pregnancy-related steroid and peptide hormones, and neuroplasticity in the brain. We discuss which brain plasticity mechanisms might underlie the structural changes detected by MRI, which hormonal systems are likely to contribute to such neuroanatomical changes and how these brain mechanisms may be linked to maternal behaviour. This Review offers an overarching framework that can serve as a roadmap for future investigations.
Collapse
Affiliation(s)
- Camila Servin-Barthet
- Unitat de Recerca en Neurociència Cognitiva, Departament de Psiquiatria i Medicina Legal, Universitat Autònoma de Barcelona, Barcelona, Spain
- Hospital del Mar Research Institute, Barcelona, Spain
| | - Magdalena Martínez-García
- Instituto de Investigación Sanitaria Gregorio Marañon, Madrid, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Clara Pretus
- Hospital del Mar Research Institute, Barcelona, Spain
- Departament de Psicobiologia i de Metodologia de els Ciències de la Salut, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria Paternina-Die
- Instituto de Investigación Sanitaria Gregorio Marañon, Madrid, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Anna Soler
- Unitat de Recerca en Neurociència Cognitiva, Departament de Psiquiatria i Medicina Legal, Universitat Autònoma de Barcelona, Barcelona, Spain
- Hospital del Mar Research Institute, Barcelona, Spain
| | | | - Óscar J Pozo
- Hospital del Mar Research Institute, Barcelona, Spain
| | - Benedetta Leuner
- Psychology Department, The Ohio State University, Columbus, OH, USA
| | - Oscar Vilarroya
- Unitat de Recerca en Neurociència Cognitiva, Departament de Psiquiatria i Medicina Legal, Universitat Autònoma de Barcelona, Barcelona, Spain.
- Hospital del Mar Research Institute, Barcelona, Spain.
| | - Susana Carmona
- Instituto de Investigación Sanitaria Gregorio Marañon, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
17
|
Powers S, Han X, Martinez J, Dufford AJ, Metz TD, Yeh T, Kim P. Cannabis use during pregnancy and hemodynamic responses to infant cues in pregnancy: an exploratory study. Front Psychiatry 2023; 14:1180947. [PMID: 37743996 PMCID: PMC10512021 DOI: 10.3389/fpsyt.2023.1180947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 08/07/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction Cannabis is one of the most commonly used substances during pregnancy and has the potential to negatively impact parent-infant relationships. The prefrontal cortex (PFC) response to infant cues during pregnancy has been associated with subsequent positive parenting behaviors. However, PFC activation is altered in individuals who use cannabis. As the potency of cannabis has changed over the years, little is known about the specific role of cannabis use on gestational parent brain responses to infant cues. Materials and methods Using functional Near-Infrared Spectroscopy (fNIRS) in the second trimester of pregnancy, we measured hemodynamic responses to an infant cry task and an infant faces task among individuals who were using cannabis (N = 14) and compared them with those who were not using cannabis (N = 45). For the infant cry task, pregnant individuals listened to cry sounds and matched white noise. For the infant faces task, they viewed happy, sad, and neutral faces. Results There was no significant difference between the two groups after adjusting for multiple comparisons. Without adjusting for multiple comparisons, we found preliminary evidence for the differences in the dorsomedial PFC associated with heightened response to infant cry among individuals who use cannabis. The groups were also different in the dorsolateral PFC associated with decreased response to infant sad faces among individuals who use cannabis. Discussion Our preliminary data suggests that cannabis use during pregnancy was associated with brain activation in the regions involved in the emotional regulation and information processes. However, the results did not survive after adjustment for multiple comparisons, thus future research with larger sample sizes is needed to confirm potential differences in brain function among cannabis-using pregnant individuals.
Collapse
Affiliation(s)
- Shannon Powers
- University of Denver, Psychology, Denver, CO, United States
| | - Xu Han
- University of Colorado, Computer Science, Boulder, CO, United States
| | | | - Alexander John Dufford
- Department of Medical Social Sciences and Institute for Innovations in Developmental Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Torri D. Metz
- University of Utah Health, Obstetrics/Gynecology, Salt Lake City, UT, United States
| | - Tom Yeh
- University of Colorado, Computer Science, Boulder, CO, United States
| | - Pilyoung Kim
- University of Denver, Psychology, Denver, CO, United States
- Department of Psychology, Ewha Womans University, Seoul, South Korea
| |
Collapse
|
18
|
Bouguiyoud N, Xie WB, Bronchti G, Frasnelli J, Al Aïn S. Enhanced maternal behaviors in a mouse model of congenital blindness. Dev Psychobiol 2023; 65:e22406. [PMID: 37607896 DOI: 10.1002/dev.22406] [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: 07/18/2022] [Revised: 05/04/2023] [Accepted: 06/07/2023] [Indexed: 08/24/2023]
Abstract
In mammals, mothering is one of the most important prosocial female behavior to promote survival, proper sensorimotor, and emotional development of the offspring. Different intrinsic and extrinsic factors can initiate and maintain these behaviors, such as hormonal, cerebral, and sensory changes. Infant cues also stimulate multisensory systems and orchestrate complex maternal responsiveness. To understand the maternal behavior driven by complex sensory interactions, it is necessary to comprehend the individual sensory systems by taking out other senses. An excellent model for investigating sensory regulation of maternal behavior is a murine model of congenital blindness, the ZRDBA mice, where both an anophthalmic and sighted mice are generated from the same litter. Therefore, this study aims to assess whether visual inputs are essential to driving maternal behaviors in mice. Maternal behaviors were assessed using three behavioral tests, including the pup retrieval test, the home cage maternal behavior test, and the maternal aggression test. Our results show that blind mothers (1) took less time to retrieve their offspring inside the nest, (2) spent more time nursing and licking their offspring in the second- and third-week postpartum, and (3) exhibited faster aggressive behaviors when exposed to an intruder male, compared to the sighted counterparts. This study provides evidence that congenitally blind mothers show more motivation to retrieve the pups, care, and protection towards their pups than sighted ones, likely due to a phenomenon of sensory compensation.
Collapse
Affiliation(s)
- Nouhaila Bouguiyoud
- Department of Anatomy, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
- CogNAC Research Group, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
| | - Wen Bin Xie
- Department of Anatomy, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
| | - Gilles Bronchti
- Department of Anatomy, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
| | - Johannes Frasnelli
- Department of Anatomy, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
- CogNAC Research Group, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
| | - Syrina Al Aïn
- Department of Anatomy, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
- CogNAC Research Group, Université du Québec à Trois-Rivières, Trois-Rivières, Quebec, Canada
| |
Collapse
|
19
|
Cheng M, Ye C, Tian C, Zhao D, Li H, Sun Z, Miao Y, Zhang Q, Wang J, Dou Y. Engineered macrophage-biomimetic versatile nanoantidotes for inflammation-targeted therapy against Alzheimer's disease by neurotoxin neutralization and immune recognition suppression. Bioact Mater 2023; 26:337-352. [PMID: 36950153 PMCID: PMC10027514 DOI: 10.1016/j.bioactmat.2023.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/21/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
Immune recognition of excessive neurotoxins by microglia is a trigger for the onset of neuroinflammation in the brain, leading to neurodegeneration in Alzheimer's disease (AD). Blocking active recognition of microglia while removing neurotoxins holds promise for fundamentally alleviating neurotoxin-induced immune responses, but is very challenging. Herein, an engineered macrophage-biomimetic versatile nanoantidote (OT-Lipo@M) is developed for inflammation-targeted therapy against AD by neurotoxin neutralization and immune recognition suppression. Coating macrophage membranes can not only endow OT-Lipo@M with anti-phagocytic and inflammation-tropism capabilities to target inflammatory lesions in AD brain, but also efficiently reduce neurotoxin levels to prevent them from activating microglia. The loaded oxytocin (OT) can be slowly released to downregulate the expression of immune recognition site Toll-like receptor 4 (TLR4) on microglia, inhibiting TLR4-mediated pro-inflammatory signalling cascade. Benefiting from this two-pronged immunosuppressive strategy, OT-Lipo@M exhibits outstanding therapeutic effects on ameliorating cognitive deficits, inhibiting neuronal apoptosis, and enhancing synaptic plasticity in AD mice, accompanied by the delayed hippocampal atrophy and brain microstructural disruption by in vivo 9.4T MR imaging. This work provides new insights into potential AD therapeutics targeting microglia-mediated neuroinflammation at the source.
Collapse
Affiliation(s)
- Meng Cheng
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Caihua Ye
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Chunxiao Tian
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, 300070, PR China
| | - Dongju Zhao
- School of Life Sciences, Tianjin University, Tianjin, 300072, PR China
| | - Haonan Li
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Zuhao Sun
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
| | - Yuyang Miao
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, 300052, PR China
| | - Qiang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, 300052, PR China
- Corresponding author.
| | - Junping Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
- Corresponding author.
| | - Yan Dou
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, PR China
- Corresponding author.
| |
Collapse
|
20
|
Luigi-Sierra MG, Guan D, López-Béjar M, Casas E, Olvera-Maneu S, Gardela J, Palomo MJ, Osuagwuh UI, Ohaneje UL, Mármol-Sánchez E, Amills M. A protein-coding gene expression atlas from the brain of pregnant and non-pregnant goats. Front Genet 2023; 14:1114749. [PMID: 37519888 PMCID: PMC10382233 DOI: 10.3389/fgene.2023.1114749] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Background: The brain is an extraordinarily complex organ with multiple anatomical structures involved in highly specialized functions related with behavior and physiological homeostasis. Our goal was to build an atlas of protein-coding gene expression in the goat brain by sequencing the transcriptomes of 12 brain regions in seven female Murciano-Granadina goats, from which three of them were 1-month pregnant. Results: Between 14,889 (cerebellar hemisphere) and 15,592 (pineal gland) protein-coding genes were expressed in goat brain regions, and most of them displayed ubiquitous or broad patterns of expression across tissues. Principal component analysis and hierarchical clustering based on the patterns of mRNA expression revealed that samples from certain brain regions tend to group according to their position in the anterior-posterior axis of the neural tube, i.e., hindbrain (pons and medulla oblongata), midbrain (rostral colliculus) and forebrain (frontal neocortex, olfactory bulb, hypothalamus, and hippocampus). Exceptions to this observation were cerebellum and glandular tissues (pineal gland and hypophysis), which showed highly divergent mRNA expression profiles. Differential expression analysis between pregnant and non-pregnant goats revealed moderate changes of mRNA expression in the frontal neocortex, hippocampus, adenohypophysis and pons, and very dramatic changes in the olfactory bulb. Many genes showing differential expression in this organ are related to olfactory function and behavior in humans. Conclusion: With the exception of cerebellum and glandular tissues, there is a relationship between the cellular origin of sampled regions along the anterior-posterior axis of the neural tube and their mRNA expression patterns in the goat adult brain. Gestation induces substantial changes in the mRNA expression of the olfactory bulb, a finding consistent with the key role of this anatomical structure on the development of maternal behavior.
Collapse
Affiliation(s)
| | - Dailu Guan
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Bellaterra, Spain
| | - Manel López-Béjar
- Department of Animal Health and Anatomy, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Encarna Casas
- Department of Animal Health and Anatomy, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Sergi Olvera-Maneu
- Department of Animal Health and Anatomy, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Jaume Gardela
- Department of Animal Health and Anatomy, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - María Jesús Palomo
- Department of Animal Medicine and Surgery, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Uchebuchi Ike Osuagwuh
- Department of Animal Medicine and Surgery, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Uchechi Linda Ohaneje
- Department of Animal Medicine and Surgery, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Emilio Mármol-Sánchez
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Bellaterra, Spain
| | - Marcel Amills
- Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Bellaterra, Spain
- Departament de Ciència Animal i dels Aliments, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
21
|
Mafrici M, Tona F, Fragiotta S, Lorenzi U, Gitto L, Toscani L. Idiopathic Intracranial Hypertension Papillopathy due to Hormonal Changes during Pregnancy. Case Rep Ophthalmol Med 2023; 2023:6688445. [PMID: 37469477 PMCID: PMC10353893 DOI: 10.1155/2023/6688445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/15/2023] [Accepted: 06/20/2023] [Indexed: 07/21/2023] Open
Abstract
Background The underlying mechanisms of papilledema associated with intracranial hypertension remain unclear. A case of bilateral papillary edema in a patient with chronic idiopathic intracranial hypertension who was asymptomatic during her two pregnancies is reported. Case Presentation. A 19-year-old Caucasian female, in her third month of pregnancy, complained of difficulties with close reading. The patient's visual acuity was 20/20 on the Snellen chart and improved with a 0.50 D correction in both eyes. Near vision and slit lamp examinations revealed normal findings bilaterally. However, a fundus examination showed bilateral papillary edema without evidence of hemorrhages or neovascularization. Blood tests were unremarkable, except for a slight increase in C-reactive protein levels. The patient had a prepregnancy weight of 63 kilograms, with a BMI of 24.91 kg/m2. Magnetic resonance imaging of the brain revealed features consistent with chronic idiopathic intracranial hypertension, which resolved after delivery. Two and a half years later, during a subsequent pregnancy, the patient experienced a recurrence of bilateral papillary edema due to the IIH. It was managed similarly as the first occurrence, resulting in bilateral anatomical and functional recovery. Recent research revealed that, during pregnancy, hormones interact with the central nervous system, leading to an increase in the size of neurons which could potentially result in intracranial hypertension. Conclusions The influence of hormonal fluctuations during pregnancy on the development of transient central nervous system abnormalities in individuals with chronic intracranial hypertension, leading to papillary edema, remains a matter of debate.
Collapse
Affiliation(s)
- Marco Mafrici
- Department of Ophthalmology, Ales-Cevennes Hospital, Ales, France
| | - Francesca Tona
- Imperial College NHS Healthcare Trust, Charing Cross Hospital, London, UK
| | - Serena Fragiotta
- NESMOS Department, Ophthalmology Unit, St. Andrea Hospital, University of Rome “La Sapienza”, Rome, Italy
| | - Umberto Lorenzi
- Department of Ophthalmology, Charles-Nicolle Hospital, Rouen, France
| | - Lorenzo Gitto
- Cook County Medical Examiner's Office, Chicago, Illinois, USA
| | - Laura Toscani
- Department of Anesthesiology and Intensive Care, CTO Andrea Alesini Hospital, Roma, Italy
| |
Collapse
|
22
|
Chotiwan N, Rosendal E, Willekens SMA, Schexnaydre E, Nilsson E, Lindqvist R, Hahn M, Mihai IS, Morini F, Zhang J, Ebel GD, Carlson LA, Henriksson J, Ahlgren U, Marcellino D, Överby AK. Type I interferon shapes brain distribution and tropism of tick-borne flavivirus. Nat Commun 2023; 14:2007. [PMID: 37037810 PMCID: PMC10086010 DOI: 10.1038/s41467-023-37698-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 03/28/2023] [Indexed: 04/12/2023] Open
Abstract
Viral tropism within the brain and the role(s) of vertebrate immune response to neurotropic flaviviruses infection is largely understudied. We combine multimodal imaging (cm-nm scale) with single nuclei RNA-sequencing to study Langat virus in wildtype and interferon alpha/beta receptor knockout (Ifnar-/-) mice to visualize viral pathogenesis and define molecular mechanisms. Whole brain viral infection is imaged by Optical Projection Tomography coregistered to ex vivo MRI. Infection is limited to grey matter of sensory systems in wildtype mice, but extends into white matter, meninges and choroid plexus in Ifnar-/- mice. Cells in wildtype display strong type I and II IFN responses, likely due to Ifnb expressing astrocytes, infiltration of macrophages and Ifng-expressing CD8+ NK cells, whereas in Ifnar-/-, the absence of this response contributes to a shift in cellular tropism towards non-activated resident microglia. Multimodal imaging-transcriptomics exemplifies a powerful way to characterize mechanisms of viral pathogenesis and tropism.
Collapse
Affiliation(s)
- Nunya Chotiwan
- Department of Clinical Microbiology, Umeå University, 90185, Umeå, Sweden.
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187, Umeå, Sweden.
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, 10540, Thailand.
| | - Ebba Rosendal
- Department of Clinical Microbiology, Umeå University, 90185, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187, Umeå, Sweden
| | - Stefanie M A Willekens
- Department of Clinical Microbiology, Umeå University, 90185, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187, Umeå, Sweden
- Umeå Centre for Molecular Medicine (UCMM), Umeå University, 90187, Umeå, Sweden
| | - Erin Schexnaydre
- Department of Clinical Microbiology, Umeå University, 90185, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, 90187, Umeå, Sweden
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187, Umeå, Sweden
- Umeå Centre for Microbial Research, Umeå University, 90187, Umeå, Sweden
| | - Emma Nilsson
- Department of Clinical Microbiology, Umeå University, 90185, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187, Umeå, Sweden
| | - Richard Lindqvist
- Department of Clinical Microbiology, Umeå University, 90185, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187, Umeå, Sweden
| | - Max Hahn
- Umeå Centre for Molecular Medicine (UCMM), Umeå University, 90187, Umeå, Sweden
| | - Ionut Sebastian Mihai
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187, Umeå, Sweden
- Department of Department of Molecular biology, Umeå University, 90187, Umeå, Sweden
- Företagsforskarskolan, Umeå University, 90187, Umeå, Sweden
| | - Federico Morini
- Umeå Centre for Molecular Medicine (UCMM), Umeå University, 90187, Umeå, Sweden
| | - Jianguo Zhang
- Department of Clinical Microbiology, Umeå University, 90185, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, 90187, Umeå, Sweden
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187, Umeå, Sweden
- Umeå Centre for Microbial Research, Umeå University, 90187, Umeå, Sweden
| | - Gregory D Ebel
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Lars-Anders Carlson
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, 90187, Umeå, Sweden
- Department of Medical Biochemistry and Biophysics, Umeå University, 90187, Umeå, Sweden
- Umeå Centre for Microbial Research, Umeå University, 90187, Umeå, Sweden
| | - Johan Henriksson
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187, Umeå, Sweden
- Umeå Centre for Microbial Research, Umeå University, 90187, Umeå, Sweden
- Department of Department of Molecular biology, Umeå University, 90187, Umeå, Sweden
| | - Ulf Ahlgren
- Umeå Centre for Molecular Medicine (UCMM), Umeå University, 90187, Umeå, Sweden
| | - Daniel Marcellino
- Department of Integrative Medical Biology, Umeå University, 90187, Umeå, Sweden
| | - Anna K Överby
- Department of Clinical Microbiology, Umeå University, 90185, Umeå, Sweden.
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, 90187, Umeå, Sweden.
| |
Collapse
|
23
|
Xue X, Zhang Y, Tao W, Wei L, Li Q, Ma T, Xu X, Wang Y, Gu X, Xu Z, Wang H, Hua Y. Longitudinal neuroplasticity after ankle sprain in mice: A voxel-based morphometry study on 11.7T MRI. J Orthop Res 2022; 41:1291-1298. [PMID: 36203347 DOI: 10.1002/jor.25458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/21/2022] [Accepted: 09/22/2022] [Indexed: 02/04/2023]
Abstract
Lateral ankle sprains (LAS) might lead to joint sensory deafferentation, which induces maladaptive neuroplasticity, especially the morphological atrophy of the cerebellar vermis. However, longitudinal evidence on the causality of injury and neural differences is still lacking. To this end, this study aimed to determine whether the morphology of the central nervous system would be altered before and after ligament transection in LAS mouse models. A total of 40 C57BL/6 mice were randomly divided among the LAS, Sham and Blank groups. We repeatedly performed the balance beam test and neural voxel-based morphometry (VBM) measurements using an 11.7 T magnetic resonance imaging before and 2 months after the surgery. The results showed that for balance outcomes, the LAS group had a significantly longer time and more slips of the balance beam tests compared with the Sham and Blank groups at 2 months after surgery, with no significant difference among the three groups before surgery. Regarding the VBM analysis, the LAS group showed significantly lower VBM values in the central lobule III of the cerebellar vermis and medial amygdalar nucleus (MEA) compared with the Sham and Blank groups after surgery, with no significant difference among the three groups before surgery. In conclusion, lateral ligament injuries might lead to morphological atrophy of the cerebellar vermis in animal models, which might pave the way for the pathological process of ankle instability after LAS.
Collapse
Affiliation(s)
- Xiao'ao Xue
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuwen Zhang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Weichu Tao
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Lei Wei
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Qianru Li
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Tengjia Ma
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoyun Xu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Yiran Wang
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xicheng Gu
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhangran Xu
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - He Wang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.,Human Phenome Institute, Fudan University, Shanghai, China.,Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence (Fudan University), Ministry of Education, Shanghai, China
| | - Yinghui Hua
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Chechko N, Dukart J, Tchaikovski S, Enzensberger C, Neuner I, Stickel S. The expectant brain-pregnancy leads to changes in brain morphology in the early postpartum period. Cereb Cortex 2022; 32:4025-4038. [PMID: 34942007 PMCID: PMC9476604 DOI: 10.1093/cercor/bhab463] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/12/2021] [Accepted: 11/15/2021] [Indexed: 11/15/2022] Open
Abstract
There is growing evidence that pregnancy may have a significant impact on the maternal brain, causing changes in its structure. To investigate the patterns of these changes, we compared nulliparous women (n = 40) with a group of primiparous women (n = 40) and multiparous mothers (n = 37) within 1-4 days postpartum, using voxel-based and surface-based morphometry (SBM). Compared with the nulliparous women, the young mothers showed decreases in gray matter volume in the bilateral hippocampus/amygdala, the orbitofrontal/subgenual prefrontal area, the right superior temporal gyrus and insula, and the cerebellum. These pregnancy-related changes in brain structure did not predict the quality of mother-infant attachment at either 3 or 12 weeks postpartum nor were they more pronounced among the multiparous women. SBM analyses showed significant cortical thinning especially in the frontal and parietal cortices, with the parietal cortical thinning likely potentiated by multiple pregnancies. We conclude that, compared with the brain of nulliparous women, the maternal brain shows widespread morphological changes shortly after childbirth. Also, the experience of pregnancy alone may not be the underlying cause of the adaptations for mothering. As regards the exact biological function of the changes in brain morphology, longitudinal research will be needed to draw any definitive conclusions.
Collapse
Affiliation(s)
- Natalia Chechko
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen, Aachen 52074, Germany
- Institute of Neuroscience and Medicine, JARA-Institute Brain Structure Function Relationship (INM 10), Research Center Jülich, Jülich 52428, Germany
- Institute of Neuroscience and Medicine, Brain & Behavior (INM-7), Research Center Jülich, Jülich 52428, Germany
| | - Jürgen Dukart
- Institute of Neuroscience and Medicine, Brain & Behavior (INM-7), Research Center Jülich, Jülich 52428, Germany
- Institute of Systems Neuroscience, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf 40225, Germany
| | - Svetlana Tchaikovski
- Department of Gynecology and Obstetrics, Medical Faculty, RWTH Aachen, Aachen 52074, Germany
| | - Christian Enzensberger
- Department of Gynecology and Obstetrics, Medical Faculty, RWTH Aachen, Aachen 52074, Germany
| | - Irene Neuner
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen, Aachen 52074, Germany
- Institute of Neuroscience and Medicine, JARA-Institute Brain Structure Function Relationship (INM 10), Research Center Jülich, Jülich 52428, Germany
| | - Susanne Stickel
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen, Aachen 52074, Germany
- Institute of Neuroscience and Medicine, JARA-Institute Brain Structure Function Relationship (INM 10), Research Center Jülich, Jülich 52428, Germany
| |
Collapse
|
25
|
Sun Z, Zhao S, Suo X, Dou Y. Sirt1 protects against hippocampal atrophy and its induced cognitive impairment in middle-aged mice. BMC Neurosci 2022; 23:33. [PMID: 35668361 PMCID: PMC9169381 DOI: 10.1186/s12868-022-00718-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 05/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Sirtuin 1 (Sirt1) is a recognized longevity gene and has been shown to be associated with aging and its related diseases. Hippocampal volume is considered to be the most sensitive brain imaging phenotype for cognition, but the effect of Sirt1 on hippocampal morphology during aging has not been reported. RESULTS Herein, we investigated the effect of conditional Sirt1 knockdown on hippocampal volume in middle-aged mice, as well as its cognitive function and the underlying molecular mechanisms. Brain structural magnetic resonance imaging (MRI) showed that adeno-associated virus (AAV) mediated hippocampal Sirt1 knockdown caused hippocampal atrophy in 8-month-old mice. Open field test (OFT) and Morris Water Maze (MWM) test revealed that hippocampal Sirt1 knockdown significantly weakened spatial learning and memory of mice without effect on anxiety and exploratory behavior. Western blotting analysis showed that P-tau levels at serine 396 epitope were significantly increased with slightly decreased T-tau levels, while PSD95 and NMDAR2B levels were obviously reduced, indicating that hippocampal Sirt1 knockdown could activate tau hyperphosphorylation and synaptic damage. CONCLUSIONS This work revealed that Sirt1 is an important protective gene against hippocampal atrophy and its induced cognitive impairment during aging, providing potential therapeutic targets for the prevention and intervention of aging-related neuropsychic diseases.
Collapse
Affiliation(s)
- Zuhao Sun
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.,School of Medical Technology, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Shuang Zhao
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
| | - Xinjun Suo
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.,School of Medical Technology, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Yan Dou
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.
| |
Collapse
|
26
|
Ye C, Cheng M, Ma L, Zhang T, Sun Z, Yu C, Wang J, Dou Y. Oxytocin Nanogels Inhibit Innate Inflammatory Response for Early Intervention in Alzheimer's Disease. ACS APPLIED MATERIALS & INTERFACES 2022; 14:21822-21835. [PMID: 35510352 DOI: 10.1021/acsami.2c00007] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Prevention of Alzheimer's disease (AD) is a global imperative, but reliable early interventions are currently lacking. Microglia-mediated chronic neuroinflammation is thought to occur in the early stage of AD and plays a critical role in AD pathogenesis. Here, oxytocin (OT)-loaded angiopep-2-modified chitosan nanogels (AOC NGs) were designed for early treatment of AD via inhibiting innate inflammatory response. Through the effective transcytosis of angiopep-2, AOC NGs were driven intravenously to cross the blood-brain barrier, enter the brain, and enrich in brain areas affected by AD. A large amount of OT was then released and specifically bound to the pathological upregulated OT receptor, thus effectively inhibiting microglial activation and reducing inflammatory cytokine levels through blocking the ERK/p38 MAPK and COX-2/iNOS NF-κB signaling pathways. Consecutive weekly intravenous administration of AOC NGs into 12-week-old young APP/PS1 mice, representing the early stage of AD, remarkably slowed the progression of Aβ deposition and neuronal apoptosis in the APP/PS1 mice as they aged and ultimately prevented cognitive impairment and delayed hippocampal atrophy. Together, the findings suggest that AOC NGs, which show good biosafety, can serve as a promising therapeutic candidate to combat neuroinflammation for early prevention of AD.
Collapse
Affiliation(s)
- Caihua Ye
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Meng Cheng
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Lin Ma
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Tianzhu Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Zuhao Sun
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Chunshui Yu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Junping Wang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Yan Dou
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| |
Collapse
|
27
|
Moog NK, Heim CM, Entringer S, Simhan HN, Wadhwa PD, Buss C. Transmission of the adverse consequences of childhood maltreatment across generations: Focus on gestational biology. Pharmacol Biochem Behav 2022; 215:173372. [DOI: 10.1016/j.pbb.2022.173372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 02/04/2022] [Accepted: 02/28/2022] [Indexed: 12/25/2022]
|
28
|
Pawluski JL, Hoekzema E, Leuner B, Lonstein JS. Less can be more: Fine tuning the maternal brain. Neurosci Biobehav Rev 2022; 133:104475. [PMID: 34864004 PMCID: PMC8807930 DOI: 10.1016/j.neubiorev.2021.11.045] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/18/2021] [Accepted: 11/30/2021] [Indexed: 02/03/2023]
Abstract
PAWLUSKI, J.L., Hoekzema, E., Leuner, B., and Lonstein, J.S. Less can be more: Fine tuning the maternal brain. NEUROSCI BIOBEHAV REV (129) XXX-XXX, 2022. Plasticity in the female brain across the lifespan has recently become a growing field of scientific inquiry. This has led to the understanding that the transition to motherhood is marked by some of the most significant changes in brain plasticity in the adult female brain. Perhaps unexpectedly, plasticity occurring in the maternal brain often involves a decrease in brain volume, neurogenesis and glial cell density that presumably optimizes caregiving and other postpartum behaviors. This review summarizes what we know of the 'fine-tuning' of the female brain that accompanies motherhood and highlights the implications of these changes for maternal neurobehavioral health. The first part of the review summarizes structural and functional brain changes in humans during pregnancy and postpartum period with the remainder of the review focusing on neural and glial plasticity during the peripartum period in animal models. The aim of this review is to provide a clear understanding of when 'less is more' in maternal brain plasticity and where future research can focus to improve our understanding of the unique brain plasticity occurring during matrescence.
Collapse
Affiliation(s)
- Jodi L. Pawluski
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France.,Corresponding author: Jodi L. Pawluski, University of Rennes 1, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000, Rennes, France.
| | - Elseline Hoekzema
- Brain and Development Laboratory, Leiden Institute for Brain and Cognition (LIBC), Leiden University, Leiden, The Netherlands.,Hoekzema Lab, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Benedetta Leuner
- The Ohio State University, Department of Psychology & Department of Neuroscience Columbus, OH, USA
| | - Joseph S. Lonstein
- Neuroscience Program & Department of Psychology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
29
|
Giacobbo BL, Özalay Ö, Mediavilla T, Ericsson M, Axelsson J, Rieckmann A, Sultan F, Marcellino D. The Aged Striatum: Evidence of Molecular and Structural Changes Using a Longitudinal Multimodal Approach in Mice. Front Aging Neurosci 2022; 14:795132. [PMID: 35140600 PMCID: PMC8818755 DOI: 10.3389/fnagi.2022.795132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/03/2022] [Indexed: 01/08/2023] Open
Abstract
To study the aging human brain requires significant resources and time. Thus, mice models of aging can provide insight into changes in brain biological functions at a fraction of the time when compared to humans. This study aims to explore changes in dopamine D1 and D2 receptor availability and of gray matter density in striatum during aging in mice and to evaluate whether longitudinal imaging in mice may serve as a model for normal brain aging to complement cross-sectional research in humans. Mice underwent repeated structural magnetic resonance imaging (sMRI), and [11C]Raclopride and [11C]SCH23390 positron emission tomography (PET) was performed on a subset of aging mice. PET and sMRI data were analyzed by binding potential (BPND), voxel- and tensor-based morphometry (VBM and TBM, respectively). Longitudinal PET revealed a significant reduction in striatal BPND for D2 receptors over time, whereas no significant change was found for D1 receptors. sMRI indicated a significant increase in modulated gray matter density (mGMD) over time in striatum, with limited clusters showing decreased mGMD. Mouse [11C]Raclopride data is compatible with previous reports in human cross-sectional studies, suggesting that a natural loss of dopaminergic D2 receptors in striatum can be assessed in mice, reflecting estimates from humans. No changes in D1 were found, which may be attributed to altered [11C]SCH23390 kinetics in anesthetized mice, suggesting that this tracer is not yet able to replicate human findings. sMRI revealed a significant increase in mGMD. Although contrary to expectations, this increase in modulated GM density may be attributed to an age-related increase in non-neuronal cells.
Collapse
Affiliation(s)
| | - Özgün Özalay
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Tomas Mediavilla
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | | | - Jan Axelsson
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Anna Rieckmann
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Fahad Sultan
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Daniel Marcellino
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
- *Correspondence: Daniel Marcellino,
| |
Collapse
|
30
|
Martínez-García M, Paternina-Die M, Desco M, Vilarroya O, Carmona S. Characterizing the Brain Structural Adaptations Across the Motherhood Transition. Front Glob Womens Health 2021; 2:742775. [PMID: 34816246 PMCID: PMC8593951 DOI: 10.3389/fgwh.2021.742775] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/10/2021] [Indexed: 12/23/2022] Open
Abstract
Women that become mothers face notable physiological adaptations during this life-period. Neuroimaging studies of the last decade have provided grounded evidence that women's brains structurally change across the transition into motherhood. The characterization of this brain remodeling is currently in its early years of research. The current article reviews this scientific field by focusing on our longitudinal (pre-to-post pregnancy) Magnetic Resonance Imaging (MRI) studies in first-time parents and other longitudinal and cross-sectional studies of parents. We present the questions that are currently being answered by the parental brain literature and point out those that have not yet been explored. We also highlight potential confounding variables that need to be considered when analyzing and interpreting brain changes observed during motherhood.
Collapse
Affiliation(s)
- Magdalena Martínez-García
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - María Paternina-Die
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| | - Manuel Desco
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Madrid, Spain.,Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Oscar Vilarroya
- Department of Psychiatry and Legal Medicine, Autonomous University of Barcelona, Barcelona, Spain.,Hospital del Mar Medical Research Institute, Barcelona, Spain
| | - Susanna Carmona
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain
| |
Collapse
|