51
|
Gjerde PB, Simonsen CE, Lagerberg TV, Steen NE, Andreassen OA, Steen VM, Melle I. Sex-Specific Effect of Serum Lipids and Body Mass Index on Psychotic Symptoms, a Cross-Sectional Study of First-Episode Psychosis Patients. Front Psychiatry 2021; 12:723158. [PMID: 34744818 PMCID: PMC8566674 DOI: 10.3389/fpsyt.2021.723158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/24/2021] [Indexed: 11/23/2022] Open
Abstract
Background: Schizophrenia is a disorder with considerable heterogeneity in course and outcomes, which is in part related to the patients' sex. Studies report a link between serum lipids, body mass index (BMI), and therapeutic response. However, the role of sex in these relationships is poorly understood. In a cross-sectional sample of first-episode psychosis (FEP) patients, we investigated if the relationship between serum lipid levels (total cholesterol, HDL-C, LDL-C, and triglycerides), BMI, and symptoms differs between the sexes. Methods: We included 435 FEP patients (males: N = 283, 65%) from the ongoing Thematically Organized Psychosis (TOP) study. Data on clinical status, antipsychotics, lifestyle, serum lipid levels, and BMI were obtained. The Positive and Negative Syndrome Scale (PANSS) and the Calgary Depression Scale for Schizophrenia (CDSS) were used to assess psychotic and depressive symptoms. General linear models were employed to examine the relationship between metabolic variables and symptomatology. Results: We observed a female-specific association between serum HDL-C levels and negative symptoms (B = -2.24, p = 0.03) and between triglycerides levels (B = 1.48, p = 0.04) and BMI (B = 0.27, p = 0.001) with depressive symptoms. When controlling for BMI, only the association between serum HDL-C levels and negative symptoms remained significant. Moreover, the HDL-C and BMI associations remained significant after controlling for demography, lifestyle, and illness-related factors. Conclusion: We found a relationship between metabolic factors and psychiatric symptoms in FEP patients that was sex-dependent.
Collapse
Affiliation(s)
- Priyanthi B Gjerde
- Norwegian Centre for Mental Disorders Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,Dr. Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway.,Research Unit for General Practice, NORCE Norwegian Research Centre, Bergen, Norway
| | - Carmen E Simonsen
- Norwegian Centre for Mental Disorders Research, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Trine V Lagerberg
- Norwegian Centre for Mental Disorders Research, Oslo University Hospital, Oslo, Norway
| | - Nils Eiel Steen
- Norwegian Centre for Mental Disorders Research, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- Norwegian Centre for Mental Disorders Research, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Vidar M Steen
- Norwegian Centre for Mental Disorders Research, Department of Clinical Science, University of Bergen, Bergen, Norway.,Dr. Einar Martens Research Group for Biological Psychiatry, Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Ingrid Melle
- Norwegian Centre for Mental Disorders Research, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
52
|
Zhou JH, Zhang DL, Ning BL, Xue XJ, Zhao L, Wu Q, Yan LD, Liu M, Fu WB. The Role of Acupuncture in Hormonal Shock-Induced Cognitive-Related Symptoms in Perimenopausal Depression: A Randomized Clinical Controlled Trial. Front Psychiatry 2021; 12:772523. [PMID: 35095593 PMCID: PMC8793332 DOI: 10.3389/fpsyt.2021.772523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/20/2021] [Indexed: 12/04/2022] Open
Abstract
Introduction: Perimenopausal depression is predominantly caused by hormone shock, but the underlying physical and psychological factors are still unclear. Objectives: To explore the constituent components of perimenopausal depression by dynamically depicting its influencing factors and interactive pathways from the perspective of embodied cognition. Methods: This is a randomized clinical controlled trial. In this study, electroacupuncture was compared with escitalopram. A total of 242 participants with mild to moderate perimenopausal depression were enrolled from 6 hospitals in China. Each participant had a 12-week intervention and a 12-week follow-up period. The primary outcome of this study is the Hamilton Depression Rating Scale (HAMD-17), and the secondary outcome is the menopause-specific quality of life scale (MENQOL), serum Follicle-stimulating hormone (FSH), luteinizing hormone (LH), and estrogen (E2) levels. Results: The structural equation model suggested that hormone levels were not directly associated with HAMD-17 (P = 0.852), while MENQOL was statistically correlated with HAMD-17 as an intermediary variable (P < 0.001). Electroacupuncture gradually showed positive impacts on MENQOL and HAMD-17 during the follow-up period (P < 0.05). Cognitive impairment is the dominant dimension of perimenopausal depression. Conclusions: Hormonal shock may affect clinical symptoms and poor quality of life to induce cognitive impairment leading perimenopausal depression, and this impact on cognition is embodied. Electroacupuncture has positive effect on perimenopausal depression and quality of life.
Collapse
Affiliation(s)
- Jun-He Zhou
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Guangzhou, China; School of Psychology Center for Studies of Psychological Application, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, China.,The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - De-Long Zhang
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Guangzhou, China; School of Psychology Center for Studies of Psychological Application, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, China
| | - Bai-Le Ning
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Guangzhou, China; School of Psychology Center for Studies of Psychological Application, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, China
| | - Xiao-Juan Xue
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Guangzhou, China; School of Psychology Center for Studies of Psychological Application, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, China
| | - Lin Zhao
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qian Wu
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lu-Da Yan
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ming Liu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education, Guangzhou, China; School of Psychology Center for Studies of Psychological Application, and Guangdong Key Laboratory of Mental Health and Cognitive Science, South China Normal University, Guangzhou, China
| | - Wen-Bin Fu
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Innovative Research Team of Acupuncture for Depression and Related Disorders, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
| |
Collapse
|
53
|
Taylor CM, Pritschet L, Olsen RK, Layher E, Santander T, Grafton ST, Jacobs EG. Progesterone shapes medial temporal lobe volume across the human menstrual cycle. Neuroimage 2020; 220:117125. [DOI: 10.1016/j.neuroimage.2020.117125] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/10/2020] [Accepted: 06/29/2020] [Indexed: 01/05/2023] Open
|
54
|
Rajizadeh MA, Esmaeilpour K, Motamedy S, Mohtashami Borzadaranb F, Sheibani V. Cognitive Impairments of Sleep-Deprived Ovariectomized (OVX) Female Rats by Voluntary Exercise. Basic Clin Neurosci 2020; 11:573-586. [PMID: 33643551 PMCID: PMC7878057 DOI: 10.32598/bcn.9.10.505] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 08/25/2019] [Accepted: 11/08/2019] [Indexed: 12/22/2022] Open
Abstract
Introduction: Previous studies demonstrated that forced and voluntary exercise had ameliorative effects on behavioral tasks followed by Sleep Deprivation (SD) in intact female rats. The main goal of this research was evaluating the impact of voluntary exercise on cognitive functions while SD and ovariectomization is induced in female wistar rats. Methods: The rats were anesthesized combining dosage of ketamine and xylazine. Then, both ovaries were eliminated and 3 weeks after surgery the animals entered the study. The exercise protocol took 4 weeks of voluntary exercise in a wheel which was connected to home cage. For inducing a 72 hours deprivation the multiple platforms was applied. The cognitive functions were studied by exploiting the Morris Water Maze (MWM) and Novel object recognition tests. Anxiety was evaluated by open field test and corticostrone measurement was carried out by ELISA method. One-way and two-way ANOVA and repeated measures were utilized for data analysis and P<0.05 was considered statistically significant. Results: We observed significant spatial and recognition learning and memory impairments in OVX sleep-deprived rats compared to the control group and voluntary exercise alleviated the SD-induced learning and memory defects. Conclusion: We concluded that voluntary exercise can improve cognitive impairments followed by SD in OVX female rats.
Collapse
Affiliation(s)
- Mohammad Amin Rajizadeh
- Department of Physiology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.,Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Khadijeh Esmaeilpour
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Sina Motamedy
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Vahid Sheibani
- Department of Physiology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.,Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
55
|
Seitz J, Cetin-Karayumak S, Lyall A, Pasternak O, Baxi M, Vangel M, Pearlson G, Tamminga C, Sweeney J, Clementz B, Schretlen D, Viher PV, Stegmayer K, Walther S, Lee J, Crow T, James A, Voineskos A, Buchanan RW, Szeszko PR, Malhotra A, Keshavan M, Koerte IK, Shenton ME, Rathi Y, Kubicki M. Investigating Sexual Dimorphism of Human White Matter in a Harmonized, Multisite Diffusion Magnetic Resonance Imaging Study. Cereb Cortex 2020; 31:201-212. [PMID: 32851404 DOI: 10.1093/cercor/bhaa220] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/08/2020] [Accepted: 06/30/2020] [Indexed: 12/17/2022] Open
Abstract
Axonal myelination and repair, critical processes for brain development, maturation, and aging, remain controlled by sexual hormones. Whether this influence is reflected in structural brain differences between sexes, and whether it can be quantified by neuroimaging, remains controversial. Diffusion-weighted magnetic resonance imaging (dMRI) is an in vivo method that can track myelination changes throughout the lifespan. We utilize a large, multisite sample of harmonized dMRI data (n = 551, age = 9-65 years, 46% females/54% males) to investigate the influence of sex on white matter (WM) structure. We model lifespan trajectories of WM using the most common dMRI measure fractional anisotropy (FA). Next, we examine the influence of both age and sex on FA variability. We estimate the overlap between male and female FA and test whether it is possible to label individual brains as male or female. Our results demonstrate regionally and spatially specific effects of sex. Sex differences are limited to limbic structures and young ages. Additionally, not only do sex differences diminish with age, but tracts within each subject become more similar to one another. Last, we show the high overlap in FA between sexes, which implies that determining sex based on WM remains open.
Collapse
Affiliation(s)
- Johanna Seitz
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, MA, Boston, 02115, USA
| | - Suheyla Cetin-Karayumak
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, MA, Boston, 02115, USA
| | - Amanda Lyall
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, MA, Boston, 02115, USA.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, MA, Boston, 02114, USA
| | - Ofer Pasternak
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, MA, Boston, 02115, USA.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, MA, Boston, 02114, USA.,Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, MA, Boston, 02115, USA
| | - Madhura Baxi
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, MA, Boston, 02115, USA.,Graduate Program of Neuroscience, Boston University, Boston, MA, 02215, USA
| | - Mark Vangel
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, MA, Boston, 02115, USA
| | - Godfrey Pearlson
- Department of Psychiatry, Yale University, New Haven, CT, 06520, USA
| | - Carol Tamminga
- Department of Psychiatry, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - John Sweeney
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, 45221, USA
| | - Brett Clementz
- Department of Psychology and Neuroscience, Bio-Imaging Research Center, University of Georgia, Athens, 30601, USA
| | - David Schretlen
- Department of Psychiatry and Behavioral Sciences, Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, 21205, USA
| | - Petra Verena Viher
- University of Bern, University Hospital of Psychiatry, Bern, 3012, Switzerland
| | - Katharina Stegmayer
- University of Bern, University Hospital of Psychiatry, Bern, 3012, Switzerland
| | - Sebastian Walther
- University of Bern, University Hospital of Psychiatry, Bern, 3012, Switzerland
| | - Jungsun Lee
- Department of Psychiatry, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 690-749, Korea
| | - Tim Crow
- Department of Psychiatry, SANE POWIC, Warneford Hospital, University of Oxford, Oxford, OX3 7 JX, UK
| | - Anthony James
- Department of Psychiatry, SANE POWIC, Warneford Hospital, University of Oxford, Oxford, OX3 7 JX, UK
| | - Aristotle Voineskos
- Center for Addiction and Mental Health, Department of Psychiatry, University of Toronto, Toronto, M5T1R8, Canada
| | - Robert W Buchanan
- Maryland Psychiatry Research Center, University of Maryland School of Medicine, Baltimore, 21228, USA
| | - Philip R Szeszko
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA.,Mental Illness Research, Education and Clinical Center, James J. Peters VA Medical Center, Bronx, New York, 10461, USA
| | - Anil Malhotra
- The Feinstein Institute for Medical Research and Zucker Hillside Hospital, Manhasset, 11030, USA
| | - Matcheri Keshavan
- Department of Psychiatry, Beth Israel Deaconess Medical Centre, Harvard Medical School, MA, Boston, 02115, USA
| | - Inga K Koerte
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, MA, Boston, 02115, USA.,cBRAIN, Department of Child and Adolescent Psychiatry, Psychosomatic and Psychotherapy, Ludwig-Maximilians-Universität, Munich, 80337, Germany
| | - Martha E Shenton
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, MA, Boston, 02115, USA.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, MA, Boston, 02114, USA.,Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, MA, Boston, 02115, USA.,VA Boston Healthcare System, Brockton, MA, 02301, USA
| | - Yogesh Rathi
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, MA, Boston, 02115, USA.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, MA, Boston, 02114, USA.,Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, MA, Boston, 02115, USA
| | - Marek Kubicki
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, MA, Boston, 02115, USA.,Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, MA, Boston, 02114, USA.,Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, MA, Boston, 02115, USA
| |
Collapse
|
56
|
Sáez-Orellana F, Octave JN, Pierrot N. Alzheimer's Disease, a Lipid Story: Involvement of Peroxisome Proliferator-Activated Receptor α. Cells 2020; 9:E1215. [PMID: 32422896 PMCID: PMC7290654 DOI: 10.3390/cells9051215] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/10/2020] [Accepted: 05/12/2020] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly. Mutations in genes encoding proteins involved in amyloid-β peptide (Aβ) production are responsible for inherited AD cases. The amyloid cascade hypothesis was proposed to explain the pathogeny. Despite the fact that Aβ is considered as the main culprit of the pathology, most clinical trials focusing on Aβ failed and suggested that earlier interventions are needed to influence the course of AD. Therefore, identifying risk factors that predispose to AD is crucial. Among them, the epsilon 4 allele of the apolipoprotein E gene that encodes the major brain lipid carrier and metabolic disorders such as obesity and type 2 diabetes were identified as AD risk factors, suggesting that abnormal lipid metabolism could influence the progression of the disease. Among lipids, fatty acids (FAs) play a fundamental role in proper brain function, including memory. Peroxisome proliferator-activated receptor α (PPARα) is a master metabolic regulator that regulates the catabolism of FA. Several studies report an essential role of PPARα in neuronal function governing synaptic plasticity and cognition. In this review, we explore the implication of lipid metabolism in AD, with a special focus on PPARα and its potential role in AD therapy.
Collapse
Affiliation(s)
- Francisco Sáez-Orellana
- Université Catholique de Louvain, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium; (F.S.-O.); (J.-N.O.)
- Institute of Neuroscience, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium
| | - Jean-Noël Octave
- Université Catholique de Louvain, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium; (F.S.-O.); (J.-N.O.)
- Institute of Neuroscience, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium
| | - Nathalie Pierrot
- Université Catholique de Louvain, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium; (F.S.-O.); (J.-N.O.)
- Institute of Neuroscience, Alzheimer Dementia, Avenue Mounier 53, SSS/IONS/CEMO-Bte B1.53.03, B-1200 Brussels, Belgium
| |
Collapse
|
57
|
Naderi M, Salahinejad A, Attaran A, Niyogi S, Chivers DP. Rapid effects of estradiol and its receptor agonists on object recognition and object placement in adult male zebrafish. Behav Brain Res 2020; 384:112514. [PMID: 32004591 DOI: 10.1016/j.bbr.2020.112514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/15/2020] [Accepted: 01/24/2020] [Indexed: 10/25/2022]
Abstract
In recent years, there has been a growing appreciation that 17β-estradiol (E2) can rapidly modulate learning and memory processes by binding to membrane estrogen receptors and cause the activation of a number of signaling cascades within the central nervous system. In this study, we sought to investigate the effects of post-training administration of E2 (100 ng/g, 1 μg/g, 10 μg/g) and involvement of the estrogen receptors (ERs) using selective ER agonists on the consolidation of object recognition (OR) and object placement memory (OP) in adult male zebrafish. The general activation of ERs with the highest E2 dose improved consolidation of memory in both learning tasks within 1.45 h of administration. Activation of classical ERs (ERα and ERβ) improved consolidation of OR memory, but had no effect on fish performance in OP task. On the other hand, activation of G protein-coupled ER1 impaired and enhanced consolidation of OR and OP memories, respectively. Memory improvement in both tasks was accompanied by a marked up-regulation in the expression of genes encoding ionotropic and metabotropic glutamate receptors in a task-dependent manner. In contrast, the down-regulation in the expression of certain ionotropic glutamate receptors was observed in fish with impaired OR memory. Moreover, our study also revealed an increase in the transcript abundance of genes associated with synaptic protein synthesis (brain-derived neurotrophic factor, synaptophysin, and the mechanistic target of rapamycin). These results suggest that E2 may affect consolidation of memory in zebrafish likely through rapid changes in synaptic morphology and function.
Collapse
Affiliation(s)
- Mohammad Naderi
- Department of Biology, University of Saskatchewan, 112 Science Place, Saskatoon, SK S7N 5E2, Canada.
| | - Arash Salahinejad
- Department of Biology, University of Saskatchewan, 112 Science Place, Saskatoon, SK S7N 5E2, Canada
| | - Anoosha Attaran
- Department of Biology, University of Saskatchewan, 112 Science Place, Saskatoon, SK S7N 5E2, Canada
| | - Som Niyogi
- Department of Biology, University of Saskatchewan, 112 Science Place, Saskatoon, SK S7N 5E2, Canada; Toxicology Centre, University of Saskatchewan, 44 Campus Drive, Saskatoon, SK S7N 5B3, Canada
| | - Douglas P Chivers
- Department of Biology, University of Saskatchewan, 112 Science Place, Saskatoon, SK S7N 5E2, Canada
| |
Collapse
|
58
|
Eid RS, Lieblich SE, Duarte-Guterman P, Chaiton JA, Mah AG, Wong SJ, Wen Y, Galea LAM. Selective activation of estrogen receptors α and β: Implications for depressive-like phenotypes in female mice exposed to chronic unpredictable stress. Horm Behav 2020; 119:104651. [PMID: 31790664 DOI: 10.1016/j.yhbeh.2019.104651] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 11/06/2019] [Accepted: 11/26/2019] [Indexed: 01/19/2023]
Abstract
The estrogen receptor (ER) mechanisms by which 17β-estradiol influences depressive-like behaviour have primarily been investigated acutely and not within an animal model of depression. Therefore, the current study aimed to dissect the contribution of ERα and ERβ to the effects of 17β-estradiol under non-stress and chronic stress conditions. Ovariectomized (OVX) or sham-operated mice were treated chronically (47 days) with 17β-estradiol (E2), the ERβ agonist diarylpropionitrile (DPN), the ERα agonist propylpyrazole-triol (PPT), or vehicle. On day 15 of treatment, mice from each group were assigned to chronic unpredictable stress (CUS; 28 days) or non-CUS conditions. Mice were assessed for anxiety- and depressive-like behaviour and hypothalamic-pituitary-adrenal (HPA) axis function. Cytokine and chemokine levels, and postsynaptic density protein 95 were measured in the hippocampus and frontal cortex, and adult hippocampal neurogenesis was assessed. Overall, the effects of CUS were more robust that those of estrogenic treatments, as seen by increased immobility in the tail suspension test (TST), reduced PSD-95 expression, reduced neurogenesis in the ventral hippocampus, and HPA axis negative feedback dysregulation. However, we also observe CUS-dependent and -independent effects of ovarian status and estrogenic treatments. The effects of CUS on PSD-95 expression, the cytokine milieu, and in TST were largely driven by PPT and DPN, indicating that these treatments were not protective. Independent of CUS, estradiol increased neurogenesis in the dorsal hippocampus, blunted the corticosterone response to an acute stressor, and increased anxiety-like behaviour. These findings provide insights into the complexities of estrogen signaling in modulating depressive-like phenotypes under non-stress and chronic stress conditions.
Collapse
Affiliation(s)
- Rand S Eid
- Graduate program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie E Lieblich
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Paula Duarte-Guterman
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Jessica A Chaiton
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Amanda G Mah
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Sarah J Wong
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Yanhua Wen
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Liisa A M Galea
- Graduate program in Neuroscience, University of British Columbia, Vancouver, BC, Canada; Department of Psychology, University of British Columbia, Vancouver, BC, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
59
|
Wang Y, Mishra A, Brinton RD. Transitions in metabolic and immune systems from pre-menopause to post-menopause: implications for age-associated neurodegenerative diseases. F1000Res 2020; 9. [PMID: 32047612 PMCID: PMC6993821 DOI: 10.12688/f1000research.21599.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2020] [Indexed: 12/13/2022] Open
Abstract
The brain undergoes two aging programs: chronological and endocrinological. This is particularly evident in the female brain, which undergoes programs of aging associated with reproductive competency. Comprehensive understanding of the dynamic metabolic and neuroinflammatory aging process in the female brain can illuminate windows of opportunities to promote healthy brain aging. Bioenergetic crisis and chronic low-grade inflammation are hallmarks of brain aging and menopause and have been implicated as a unifying factor causally connecting genetic risk factors for Alzheimer's disease and other neurodegenerative diseases. In this review, we discuss metabolic phenotypes of pre-menopausal, peri-menopausal, and post-menopausal aging and their consequent impact on the neuroinflammatory profile during each transition state. A critical aspect of the aging process is the dynamic metabolic neuro-inflammatory profiles that emerge during chronological and endocrinological aging. These dynamic systems of biology are relevant to multiple age-associated neurodegenerative diseases and provide a therapeutic framework for prevention and delay of neurodegenerative diseases of aging. While these findings are based on investigations of the female brain, they have a broader fundamental systems of biology strategy for investigating the aging male brain. Molecular characterization of alterations in fuel utilization and neuroinflammatory mechanisms during these neuro-endocrine transition states can inform therapeutic strategies to mitigate the risk of Alzheimer's disease in women. We further discuss a precision hormone replacement therapy approach to target symptom profiles during endocrine and chronological aging to reduce risk for age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Yiwei Wang
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, 85721, USA
| | - Aarti Mishra
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, 85721, USA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
60
|
Yare K, Woodward M. Hormone Therapy and Effects on Sporadic Alzheimer’s Disease in Postmenopausal Women: Importance of Nomenclature. J Alzheimers Dis 2020; 73:23-37. [DOI: 10.3233/jad-190896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Katrine Yare
- Austin Health, Heidelberg Repatriation Hospital, Victoria, Australia
| | - Michael Woodward
- Austin Health, Heidelberg Repatriation Hospital, Victoria, Australia
| |
Collapse
|
61
|
Pompili A, Iorio C, Gasbarri A. Effects of sex steroid hormones on memory. Acta Neurobiol Exp (Wars) 2020. [DOI: 10.21307/ane-2020-012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
62
|
Wang T, Yao J, Chen S, Mao Z, Brinton RD. Allopregnanolone Reverses Bioenergetic Deficits in Female Triple Transgenic Alzheimer's Mouse Model. Neurotherapeutics 2020; 17:178-188. [PMID: 31664643 PMCID: PMC7053503 DOI: 10.1007/s13311-019-00793-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Previously, we reported that the neurosteroid allopregnanolone (Allo) promoted neural stem cell regeneration, restored cognitive function, and reduced Alzheimer's Disease (AD) pathology in the triple transgenic Alzheimer's mouse model (3xTgAD). To investigate the underlying systems biology of Allo action in AD models in vivo, we assessed the regulation of Allo on the bioenergetic system of the brain. Outcomes of these analysis indicated that Allo significantly reversed deficits in mitochondrial respiration and biogenesis and key mitochondrial enzyme activity and reduced lipid peroxidation in the 3xTgAD mice in vivo. To explore the mechanisms by which Allo regulates the brain metabolism, we conducted targeted transcriptome analysis. These data further confirmed that Allo upregulated genes involved in glucose metabolism, mitochondrial bioenergetics, and signaling pathways while simultaneously downregulating genes involved in Alzheimer's pathology, fatty acid metabolism, and mitochondrial uncoupling and dynamics. Upstream regulatory pathway analysis predicted that Allo induced peroxisome proliferator-activated receptor gamma (PPARG) and coactivator 1-alpha (PPARGC1A) pathways while simultaneously inhibiting the presenilin 1 (PSEN 1), phosphatase and tensin homolog (PTEN), and tumor necrosis factor (TNF) pathways to reduce AD pathology. Collectively, these data indicate that Allo functions as a systems biology regulator of bioenergetics, cholesterol homeostasis, and β-amyloid reduction in the brain. These systems are critical to neurological health, thus providing a plausible mechanistic rationale for Allo as a therapeutic to promote neural cell function and reduce the burden of AD pathology.
Collapse
Affiliation(s)
- Tian Wang
- Center for Innovation in Brain Science, University of Arizona, 1230 N Cherry Avenue, Tucson, AZ, 85721, United States
| | - Jia Yao
- Department of Pharmacology and Pharmaceutical Science, School of Pharmacy, University of Southern California, California, Los Angeles, USA
| | - Shuhua Chen
- Center for Innovation in Brain Science, University of Arizona, 1230 N Cherry Avenue, Tucson, AZ, 85721, United States
| | - Zisu Mao
- Center for Innovation in Brain Science, University of Arizona, 1230 N Cherry Avenue, Tucson, AZ, 85721, United States
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, University of Arizona, 1230 N Cherry Avenue, Tucson, AZ, 85721, United States.
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona, USA.
- Department of Neurology, College of Medicine, University of Arizona, Tucson, Arizona, USA.
| |
Collapse
|
63
|
Wang Y, Hernandez G, Mack WJ, Schneider LS, Yin F, Brinton RD. Retrospective analysis of phytoSERM for management of menopause-associated vasomotor symptoms and cognitive decline: a pilot study on pharmacogenomic effects of mitochondrial haplogroup and APOE genotype on therapeutic efficacy. Menopause 2020; 27:57-65. [PMID: 31567873 PMCID: PMC7100617 DOI: 10.1097/gme.0000000000001418] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE PhytoSERM is a selective estrogen receptor beta (ERβ) modulator comprised of three phytoestrogens: genistein, daidzein, and S-equol. The PhytoSERM formulation promotes estrogenic action in the brain while largely inactive or inhibitory in reproductive tissue. A phase Ib/IIa clinical trial (ClinicalTrial.gov ID: NCT01723917) of PhytoSERM demonstrated safety and pharmacokinetics profile of PhytoSERM. While this study was not powered for efficacy analysis, we conducted a pilot, retrospective analysis to identify potential responders to PhytoSERM treatment, and to determine the optimal populations to pursue in a phase II clinical trial of efficacy of the PhytoSERM formulation. METHODS In this retrospective analysis involving 46 participants (n = 16, placebo; n = 18, 50 mg/d PhytoSERM; and n = 12, 100 mg/d PhytoSERM), the therapeutic effect of PhytoSERM was stratified by 2 genetic risk modulators for Alzheimer's disease: mitochondrial haplogroup and APOE genotype. RESULTS Our retrospective responder analysis indicated that participants on 50 mg of daily PhytoSERM (PS50) for 12 weeks significantly reduced hot flash frequency compared with their baseline (mean [95% CI])-1.61, [-2.79, -0.42], P = 0.007). Participants on 50 mg of PhytoSERM also had significantly greater reduction in hot flash frequency at 12 weeks compared with the placebo group (-1.38, -0.17 [median PS50, median placebo], P = 0.04). Fifty milligrams of daily PhytoSERM also preserved cognitive function in certain aspects of verbal learning and executive function. Our analysis further suggests that mitochondrial haplogroup and APOE genotype can modify PhytoSERM response. CONCLUSION Our data support a precision medicine approach for further development of PhytoSERM as a safe and effective alternative to hormone therapy for menopause-associated hot flash and cognitive decline. While definitive determination of PhytoSERM efficacy is limited by the small sample size, these data provide a reasonable rationale to extend analyses to a larger study set powered to address statistical significance.
Collapse
Affiliation(s)
- Yiwei Wang
- School of Pharmacy, University of Southern California, Los Angeles, CA
- Center for Innovation in Brain Science and Department of Pharmacology, University of Arizona, Tucson, AZ
| | - Gerson Hernandez
- School of Pharmacy, University of Southern California, Los Angeles, CA
- Center for Innovation in Brain Science and Department of Pharmacology, University of Arizona, Tucson, AZ
| | - Wendy J Mack
- Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Lon S Schneider
- Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Fei Yin
- School of Pharmacy, University of Southern California, Los Angeles, CA
- Center for Innovation in Brain Science and Department of Pharmacology, University of Arizona, Tucson, AZ
| | - Roberta D Brinton
- School of Pharmacy, University of Southern California, Los Angeles, CA
- Center for Innovation in Brain Science and Department of Pharmacology, University of Arizona, Tucson, AZ
| |
Collapse
|
64
|
Konishi K, Cherkerzian S, Aroner S, Jacobs EG, Rentz DM, Remington A, Aizley H, Hornig M, Klibanski A, Goldstein JM. Impact of BDNF and sex on maintaining intact memory function in early midlife. Neurobiol Aging 2019; 88:137-149. [PMID: 31948671 DOI: 10.1016/j.neurobiolaging.2019.12.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 10/30/2019] [Accepted: 12/14/2019] [Indexed: 01/06/2023]
Abstract
Sex steroid hormones and neurotrophic factors, such as brain-derived neurotrophic factor (BDNF), play a significant neuroprotective role in memory circuitry aging. Here, we present findings characterizing the neuroprotective effects of BDNF on memory performance, as a function of sex and reproductive status in women. Participants (N = 191; mean age = 50.03 ± 2.10) underwent clinical and cognitive testing, fMRI scanning, and hormonal assessments of menopausal staging. Memory performance was assessed with the 6-Trial Selective Reminding Test and the Face-Name Associative Memory Exam. Participants also performed a working memory (WM) N-back task during fMRI scanning. Results revealed significant interactions between menopausal status and BDNF levels. Only in postmenopausal women, lower plasma BDNF levels were associated with significantly worse memory performance and altered function in the WM circuitry. BDNF had no significant impact on memory performance or WM function in pre/perimenopausal women or men. These results suggest that in postmenopausal women, BDNF is associated with memory performance and memory circuitry function, thus providing evidence of potential sex-dependent factors of risk and resilience for early intervention.
Collapse
Affiliation(s)
- Kyoko Konishi
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sara Cherkerzian
- Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Sarah Aroner
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Emily G Jacobs
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, CA, USA
| | - Dorene M Rentz
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anne Remington
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Harlyn Aizley
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mady Hornig
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Anne Klibanski
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jill M Goldstein
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Division of Women's Health, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
65
|
Rahman A, Jackson H, Hristov H, Isaacson RS, Saif N, Shetty T, Etingin O, Henchcliffe C, Brinton RD, Mosconi L. Sex and Gender Driven Modifiers of Alzheimer's: The Role for Estrogenic Control Across Age, Race, Medical, and Lifestyle Risks. Front Aging Neurosci 2019; 11:315. [PMID: 31803046 PMCID: PMC6872493 DOI: 10.3389/fnagi.2019.00315] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 10/31/2019] [Indexed: 12/22/2022] Open
Abstract
Research indicates that after advanced age, the major risk factor for late-onset Alzheimer’s disease (AD) is female sex. Out of every three AD patients, two are females with postmenopausal women contributing to over 60% of all those affected. Sex- and gender-related differences in AD have been widely researched and several emerging lines of evidence point to different vulnerabilities that contribute to dementia risk. Among those being considered, it is becoming widely accepted that gonadal steroids contribute to the gender disparity in AD, as evidenced by the “estrogen hypothesis.” This posits that sex hormones, 17β-estradiol in particular, exert a neuroprotective effect by shielding females’ brains from disease development. This theory is further supported by recent findings that the onset of menopause is associated with the emergence of AD-related brain changes in women in contrast to men of the same age. In this review, we discuss genetic, medical, societal, and lifestyle risk factors known to increase AD risk differently between the genders, with a focus on the role of hormonal changes, particularly declines in 17β-estradiol during the menopause transition (MT) as key underlying mechanisms.
Collapse
Affiliation(s)
- Aneela Rahman
- Department of Neurology, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Hande Jackson
- Department of Neurology, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Hollie Hristov
- Department of Neurology, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Richard S Isaacson
- Department of Neurology, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Nabeel Saif
- Department of Neurology, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Teena Shetty
- Concussion Clinic, Hospital for Special Surgery, New York, NY, United States
| | - Orli Etingin
- Department of Internal Medicine, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Claire Henchcliffe
- Department of Neurology, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Roberta Diaz Brinton
- Department of Pharmacology, College of Medicine, The University of Arizona, Tucson, AZ, United States.,Department of Neurology, College of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Lisa Mosconi
- Department of Neurology, Weill Cornell Medicine, Cornell University, New York, NY, United States.,Department of Radiology, Weill Cornell Medicine, Cornell University, New York, NY, United States.,Department of Psychiatry, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
66
|
Impact of quercetin on tight junctional proteins and BDNF signaling molecules in hippocampus of PCBs-exposed rats. Interdiscip Toxicol 2019; 11:294-305. [PMID: 31762681 PMCID: PMC6853011 DOI: 10.2478/intox-2018-0029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 02/06/2018] [Indexed: 12/26/2022] Open
Abstract
Polychlorinated biphenyls (PCBs) consist of a range of toxic substances which are directly proportional to carcinogenesis and tumor-promoting factors as well as having neurotoxic properties. Reactive oxygen species, which are produced from PCBs, alter blood–brain barrier (BBB) integrity, which is paralleled by cytoskeletal rearrangements and redistribution and disappearance of tight junction proteins (TJPs) like claudin-5 and occludin. Brain-derived neurotrophic factor (BDNF), plays an important role in the maintenance, survival of neurons and synaptic plasticity. It is predominant in the hippocampal areas vital to learning, memory and higher thinking. Quercetin, a flavonoid, had drawn attention to its neurodefensive property. The study is to assess the role of quercetin on serum PCB, estradiol and testosterone levels and mRNA expressions of estrogen receptor α and β, TJPs and BDNF signaling molecules on the hippocampus of PCBs-exposed rats. Rats were divided into 4 groups of 6 each. Group I rats were intraperitoneally (i.p.) administered corn oil (vehicle). Group II received quercetin 50 mg/kg/bwt (gavage). Group III received PCBs (Aroclor 1254) at 2 mg/kg bwt (i.p). Group IV received quercetin 50 mg/kg bwt (gavage) simultaneously with PCBs 2 mg/kg bwt (i.p.). The treatment was given daily for 30 days. The rats were euthanized 24 h after the experimental period. Blood was collected for quantification of serum PCBs estradiol and testosterone. The hippocampus was dissected and processed for PCR and Western blot; serum PCB was observed in PCB treated animals, simultaneously quercetin treated animals showed PCB metabolites. Serum testosterone and estradiol were decreased after PCB exposure. Quercetin supplementation brought back normal levels. mRNA expressions of estrogen α and β were decreased in the hippocampus of PCB treated rats. TJPS and BDNF signalling molecules were decreased in hippocampus of PCB treated rats. Quercetin supplementation retrieved all the parameters. Quercetin alone treated animals showed no alteration. Thus in PCB caused neurotoxicity, quercetin protects and prevents neuronal damage in the hippocampus.
Collapse
|
67
|
Female reproductive factors and risk of external causes of death among women: The Japan Public Health Center-based Prospective Study (JPHC Study). Sci Rep 2019; 9:14329. [PMID: 31586153 PMCID: PMC6778214 DOI: 10.1038/s41598-019-50890-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 09/16/2019] [Indexed: 01/10/2023] Open
Abstract
Although empirical data suggest a possible link between female reproductive events and risk of nonfatal accidents and suicidal behaviors, evidence to determine these effects on mortality is scarce. This study investigated the association between female reproductive factors and the risk of external causes of death among middle-aged Japanese women. We used a population-based cohort study consisting of 71 698 women residing in 11 public health center areas across Japan between 1990 and 1994. Multivariable-adjusted Cox proportional hazard regression models were used to estimate hazard ratios (HRs) of the risk of all external causes, suicide, and accidents according to female reproductive factors at the baseline survey. During 1 028 583 person-years of follow-up for 49 279 eligible subjects (average 20.9 years), we identified 328 deaths by all injuries. Among parous women, ever versus never breastfeeding [0.67 (95% CI: 0.49–0.92)] was associated with a decreased risk of all injuries. Risk of suicide was inversely associated with ever versus never parity [0.53 (95% CI: 0.32–0.88)]. A lower risk of death by accidents was seen in ever breastfeeding [0.63 (95% CI: 0.40–0.97)] compared to never breastfeeding. This study suggests that parity and breastfeeding are associated with reduced risk of death by all external causes, suicide and/or accidents among Japanese women.
Collapse
|
68
|
Chung SW, Thomson CJ, Lee S, Worsley RN, Rogasch NC, Kulkarni J, Thomson RH, Fitzgerald PB, Segrave RA. The influence of endogenous estrogen on high-frequency prefrontal transcranial magnetic stimulation. Brain Stimul 2019; 12:1271-1279. [DOI: 10.1016/j.brs.2019.05.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/03/2019] [Accepted: 05/06/2019] [Indexed: 01/06/2023] Open
|
69
|
Sadrtdinova II, Khizmatullina ZR. [Reactive changes in morphological and morphometric parameters of immunopositive astrocytes of the amygdala in response to hormone level in absence epilepsy]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 118:61-66. [PMID: 30698546 DOI: 10.17116/jnevro201811810261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
AIM To study the changes in the morphological and morphometric parameters of immunopositive astrocytes of the amygdala in absence epilepsy depending on hormonal profile. MATERIAL AND METHODS Adult female WAG/Rij rats were used as experimental subjects. The astrocytes were detected on serial sections using a reaction to glial fibrillary acidic protein (GFAP) with pre-stained hematoxylin. Quantitative analysis was carried out for a 204.8´153.6 μm2 field of view. RESULTS In the control group, astrocytes had a relatively regular stellate form and GFAP was moderately expressed in their bodies and processes. The number of astrocytes was 18.20±2.87, and their area was 164±3.29 μm2. After ovariectomy, a high expression of the protein, both in the bodies and in the processes of astrocytes, increasing the cell size to 188.85±4.97 μm2 (p<0.05) was observed. The astrocytes increased to 34.55±3.03 (p<0.05). In addition, the deformation of the processes and their diffuse defibration were observed. After hormone replacement therapy, a decrease in GFAP expression was detected, the area of astrocytes became smaller in comparison with the group after ovariectomy: 173.54±5.48 μm2 (p<0.05). Morphological changes in glial cells were manifested as a decrease in the size of their bodies, the processes became smooth without diffuse sprouting and swelling, which is probably associated with neuroprotective functions of estradiol. CONCLUSION Thus, the results of our study demonstrated that the deficiency of female sex hormones led to the increase in both the amount and area of astrocytes in the anterior cortical nucleus of the amygdala, and hormone replacement therapy positively affected the structural and quantitative characteristics of astrocytes due to the endogenous protective role of estradiol.
Collapse
|
70
|
Crimins JL, Puri R, Calakos KC, Yuk F, Janssen WGM, Hara Y, Rapp PR, Morrison JH. Synaptic distributions of pS214-tau in rhesus monkey prefrontal cortex are associated with spine density, but not with cognitive decline. J Comp Neurol 2019; 527:856-873. [PMID: 30408169 PMCID: PMC6333519 DOI: 10.1002/cne.24576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/18/2018] [Accepted: 10/21/2018] [Indexed: 12/31/2022]
Abstract
Female rhesus monkeys and women are subject to age- and menopause-related deficits in working memory, an executive function mediated by the dorsolateral prefrontal cortex (dlPFC). Long-term cyclic administration of 17β-estradiol improves working memory, and restores highly plastic axospinous synapses within layer III dlPFC of aged ovariectomized monkeys. In this study, we tested the hypothesis that synaptic distributions of tau protein phosphorylated at serine 214 (pS214-tau) are altered with age or estradiol treatment, and couple to working memory performance. First, ovariectormized young and aged monkeys received vehicle or estradiol treatment, and were tested on the delayed response (DR) test of working memory. Serial section electron microscopic immunocytochemistry was then performed to quantitatively assess the subcellular synaptic distributions of pS214-tau. Overall, the majority of synapses contained pS214-tau immunogold particles, which were predominantly localized to the cytoplasm of axon terminals. pS214-tau was also abundant within synaptic and cytoplasmic domains of dendritic spines. The density of pS214-tau immunogold within the active zone, cytoplasmic, and plasmalemmal domains of axon terminals, and subjacent to the postsynaptic density within the subsynaptic domains of dendritic spines, were each reduced with age. None of the variables examined were directly linked to cognitive status, but a high density of pS214-tau immunogold particles within presynaptic cytoplasmic and plasmalemmal domains, and within postsynaptic subsynaptic and plasmalemmal domains, accompanied high synapse density. Together, these data support a possible physiological, rather than pathological, role for pS214-tau in the modulation of synaptic morphology in monkey dlPFC.
Collapse
Affiliation(s)
- Johanna L. Crimins
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Rishi Puri
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Katina C. Calakos
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Frank Yuk
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - William G. M. Janssen
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Yuko Hara
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Peter R. Rapp
- National Institute on Aging, Laboratory of Behavioral Neuroscience, Baltimore, MD 21224
| | - John H. Morrison
- Fishberg Department of Neuroscience and Kastor Neurobiology of Aging Laboratories Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- California National Primate Research Center, Davis, CA 95616
- Department of Neurology, School of Medicine, University of California, Davis, CA 95616
| |
Collapse
|
71
|
Medeiros ADM, Silva RH. Sex Differences in Alzheimer’s Disease: Where Do We Stand? J Alzheimers Dis 2019; 67:35-60. [DOI: 10.3233/jad-180213] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- André de Macêdo Medeiros
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
- Center of Health and Biological Sciences, Universidade Federal Rural do Semiárido, Mossoró, Brazil
| | - Regina Helena Silva
- Behavioral Neuroscience Laboratory, Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
72
|
Seitz J, Kubicki M, Jacobs EG, Cherkerzian S, Weiss BK, Papadimitriou G, Mouradian P, Buka S, Goldstein JM, Makris N. Impact of sex and reproductive status on memory circuitry structure and function in early midlife using structural covariance analysis. Hum Brain Mapp 2018; 40:1221-1233. [PMID: 30548738 DOI: 10.1002/hbm.24441] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 10/11/2018] [Accepted: 10/13/2018] [Indexed: 01/13/2023] Open
Abstract
Research on age-related memory alterations traditionally targets individuals aged ≥65 years. However, recent studies emphasize the importance of early aging processes. We therefore aimed to characterize variation in brain gray matter structure in early midlife as a function of sex and menopausal status. Subjects included 94 women (33 premenopausal, 29 perimenopausal, and 32 postmenopausal) and 99 demographically comparable men from the New England Family Study. Subjects were scanned with a high-resolution T1 sequence on a 3 T whole body scanner. Sex and reproductive-dependent structural differences were evaluated using Box's M test and analysis of covariances (ANCOVAs) for gray matter volumes. Brain regions of interest included dorsolateral prefrontal cortex (DLPFC), inferior parietal lobule (iPAR), anterior cingulate cortex (ACC), hippocampus (HIPP), and parahippocampus. While we observed expected significant sex differences in volume of hippocampus with women of all groups having higher volumes than men relative to cerebrum size, we also found significant differences in the covariance matrices of perimenopausal women compared with postmenopausal women. Associations between ACC and HIPP/iPAR/DLPFC were higher in postmenopausal women and correlated with better memory performance. Findings in this study underscore the importance of sex and reproductive status in early midlife for understanding memory function with aging.
Collapse
Affiliation(s)
- Johanna Seitz
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Marek Kubicki
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Departments of Psychiatry, Neurology and Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Center for Morphometric Analysis, Center for Neural Systems Investigations, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,Department of Psychiatry, Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Emily G Jacobs
- Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Division of Women's Health, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sara Cherkerzian
- Department of Psychiatry, Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Division of Women's Health, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Blair K Weiss
- Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Division of Women's Health, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - George Papadimitriou
- Departments of Psychiatry, Neurology and Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Center for Morphometric Analysis, Center for Neural Systems Investigations, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Palig Mouradian
- Departments of Psychiatry, Neurology and Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Center for Morphometric Analysis, Center for Neural Systems Investigations, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| | - Stephen Buka
- Department of Community Health, Brown University, Providence, Rhode Island
| | - Jill M Goldstein
- Departments of Psychiatry, Neurology and Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Center for Morphometric Analysis, Center for Neural Systems Investigations, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,Department of Psychiatry, Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts.,Department of Medicine, Harvard Medical School, Boston, Massachusetts.,Division of Women's Health, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nikos Makris
- Psychiatry Neuroimaging Laboratory, Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.,Departments of Psychiatry, Neurology and Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Center for Morphometric Analysis, Center for Neural Systems Investigations, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts
| |
Collapse
|
73
|
Marchese E, Corvino V, Di Maria V, Furno A, Giannetti S, Cesari E, Lulli P, Michetti F, Geloso MC. The Neuroprotective Effects of 17β-Estradiol Pretreatment in a Model of Neonatal Hippocampal Injury Induced by Trimethyltin. Front Cell Neurosci 2018; 12:385. [PMID: 30416427 PMCID: PMC6213803 DOI: 10.3389/fncel.2018.00385] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 10/08/2018] [Indexed: 12/12/2022] Open
Abstract
Hippocampal dysfunction plays a central role in neurodevelopmental disorders, resulting in severe impairment of cognitive abilities, including memory and learning. On this basis, developmental studies represent an important tool both to understanding the cellular and molecular phenomena underlying early hippocampal damage and to study possible therapeutic interventions, that may modify the progression of neuronal death. Given the modulatory role played by 17β-estradiol (E2) on hippocampal functions and its neuroprotective properties, the present study investigates the effects of pretreatment with E2 in a model of neonatal hippocampal injury obtained by trimethyltin (TMT) administration, characterized by neuronal loss in CA1 and CA3 subfields and astroglial and microglial activation. At post-natal days (P)5 and P6 animals received E2 administration (0.2 mg/kg/die i.p.) or vehicle. At P7 they received a single dose of TMT (6.5 mg/kg i.p.) and were sacrificed 72 h (P10) or 7 days after TMT treatment (P14). Our findings indicate that pretreatment with E2 exerts a protective effect against hippocampal damage induced by TMT administration early in development, reducing the extent of neuronal death in the CA1 subfield, inducing the activation of genes involved in neuroprotection, lowering the neuroinflammatory response and restoring neuropeptide Y- and parvalbumin- expression, which is impaired in the early phases of TMT-induced damage. Our data support the efficacy of estrogen-based neuroprotective approaches to counteract early occurring hippocampal damage in the developing hippocampus.
Collapse
Affiliation(s)
- Elisa Marchese
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Corvino
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Di Maria
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Epilepsy Center, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Alfredo Furno
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Stefano Giannetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Eleonora Cesari
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Laboratory of Neuroembryology, Fondazione Santa Lucia, Rome, Italy
| | - Paola Lulli
- Laboratorio di Biochimica Clinica e Biologia Molecolare, IRCCS Fondazione Policlinico A. Gemelli, Rome, Italy
| | - Fabrizio Michetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy.,Facoltà di Medicina e Chirurgia - IRCCS San Raffaele Scientific Institute, Università Vita-Salute San Raffaele, Milan, Italy
| | - Maria Concetta Geloso
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
74
|
Abstract
During the last decades, research on adipose tissues has spread in parallel with the extension of obesity. Several observations converged on the idea that adipose tissues are organized in a large organ with endocrine and plastic properties. Two parenchymal components: white (WATs) and brown adipose tissues (BATs) are contained in subcutaneous and visceral compartments. Although both have endocrine properties, their function differs: WAT store lipids to allow intervals between meals, BAT burns lipids for thermogenesis. In spite of these opposite functions, they share the ability for reciprocal reversible transdifferentiation to tackle special physiologic needs. Thus, chronic need for thermogenesis induces browning and chronic positive energy balance induce whitening. Lineage tracing and data from explant studies strongly suggest other remodeling properties of this organ. During pregnancy and lactation breast WAT transdifferentiates into milk-secreting glands, composed by cells with abundant cytoplasmic lipids (pink adipocytes) and in the postlactation period pink adipocytes transdifferentiate back into WAT and BAT. The plastic properties of mature adipocytes are supported also by a liposecretion process in vitro where adult cell in culture transdifferentiate to differentiated fibroblast-like elements able to give rise to different phenotypes (rainbow adipocytes). In addition, the inflammasome system is activated in stressed adipocytes from obese adipose tissue. These adipocytes die and debris are reabsorbed by macrophages inducing a chronic low-grade inflammation, potentially contributing to insulin resistance and T2 diabetes. Thus, the plastic properties of this organ could open new therapeutic perspectives in the obesity-related metabolic disease and in breast pathologies. © 2018 American Physiological Society. Compr Physiol 8:1357-1431, 2018.
Collapse
Affiliation(s)
- Saverio Cinti
- Professor of Human Anatomy, Director, Center of Obesity, University of Ancona (Politecnica delle Marche), Ancona, Italy
| |
Collapse
|
75
|
Abstract
Contribution to Special Issue on Fast effects of steroids. The concept that the positive feedback effect of ovarian estradiol (E2) results in GnRH and gonadotropin surges is a well-established principle. However, a series of studies investigating the rapid action of E2 in female rhesus monkeys has led to a new concept that neuroestradiol, synthesized and released in the hypothalamus, also contributes to regulation of the preovulatory GnRH surge. This unexpected finding started from our surprising observation that E2 induces rapid stimulatory action in GnRH neurons in vitro. Subsequently, we confirmed that a similar rapid stimulatory action of E2 occurs in vivo. Unlike subcutaneous injection of E2 benzoate (EB), a brief (10-20 min), direct infusion of EB into the median eminence in ovariectomized (OVX) female monkeys rapidly stimulates release of GnRH and E2 in a pulsatile manner, and the EB-induced GnRH and E2 release is blocked by simultaneous infusion of the aromatase inhibitor, letrozole. This suggests that stimulated release of E2 is of hypothalamic origin. To further determine the role of neuroestradiol we examined the effects of letrozole on EB-induced GnRH and LH surges in OVX females. Results indicate that letrozole treatment greatly attenuated the EB-induced GnRH and LH surges. Collectively, neuroestradiol released from the hypothalamus appears to be necessary for the positive feedback effect of E2 on the GnRH/LH surge.
Collapse
Affiliation(s)
- Ei Terasawa
- Wisconsin National Primate Research Center, University of Wisconsin, Madison, WI 53715, United States; Department of Pediatrics, University of Wisconsin, Madison, WI 53706, United States.
| |
Collapse
|
76
|
Estrogen Treatment Reverses Prematurity-Induced Disruption in Cortical Interneuron Population. J Neurosci 2018; 38:7378-7391. [PMID: 30037831 DOI: 10.1523/jneurosci.0478-18.2018] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Revised: 05/22/2018] [Accepted: 06/17/2018] [Indexed: 01/21/2023] Open
Abstract
Development of cortical interneurons continues until the end of human pregnancy. Premature birth deprives the newborns from the supply of maternal estrogen and a secure intrauterine environment. Indeed, preterm infants suffer from neurobehavioral disorders. This can result from both preterm birth and associated postnatal complications, which might disrupt recruitment and maturation of cortical interneurons. We hypothesized that interneuron subtypes, including parvalbumin-positive (PV+), somatostatin-positive (SST+), calretinin-positive (CalR+), and neuropeptide Y-positive (NPY+) interneurons, were recruited in the upper and lower cortical layers in a distinct manner with advancing gestational age. In addition, preterm birth would disrupt the heterogeneity of cortical interneurons, which might be reversed by estrogen treatment. These hypotheses were tested by analyzing autopsy samples from premature infants and evaluating the effect of estrogen supplementation in prematurely delivered rabbits. The PV+ and CalR+ neurons were abundant, whereas SST+ and NPY+ neurons were few in cortical layers of preterm human infants. Premature birth of infants reduced the density of PV+ or GAD67+ neurons and increased SST+ interneurons in the upper cortical layers. Importantly, 17 β-estradiol treatment in preterm rabbits increased the number of PV+ neurons in the upper cortical layers relative to controls at postnatal day 14 (P14) and P21 and transiently reduced SST population at P14. Moreover, protein and mRNA levels of Arx, a key regulator of cortical interneuron maturation and migration, were higher in estrogen-treated rabbits relative to controls. Therefore, deficits in PV+ and excess of SST+ neurons in premature newborns are ameliorated by estrogen replacement, which can be attributed to elevated Arx levels. Estrogen replacement might enhance neurodevelopmental outcomes in extremely preterm infants.SIGNIFICANCE STATEMENT Premature birth often leads to neurodevelopmental delays and behavioral disorders, which may be ascribed to disturbances in the development and maturation of cortical interneurons. Here, we show that preterm birth in humans is associated with reduced population of parvalbumin-positive (PV+) neurons and an excess of somatostatin-expressing interneurons in the cerebral cortex. More importantly, 17 β-estradiol treatment increased the number of PV+ neurons in preterm-born rabbits, which appears to be mediated by an elevation in the expression of Arx transcription factor. Hence the present study highlights prematurity-induced reduction in PV+ neurons in human infants and reversal in their population by estrogen replacement in preterm rabbits. Because preterm birth drops plasma estrogen level 100-fold, estrogen replacement in extremely preterm infants might improve their developmental outcome and minimize neurobehavioral disorders.
Collapse
|
77
|
Mantor D, Pratchayasakul W, Minta W, Sutham W, Palee S, Sripetchwandee J, Kerdphoo S, Jaiwongkum T, Sriwichaiin S, Krintratun W, Chattipakorn N, Chattipakorn SC. Both oophorectomy and obesity impaired solely hippocampal-dependent memory via increased hippocampal dysfunction. Exp Gerontol 2018; 108:149-158. [PMID: 29678475 DOI: 10.1016/j.exger.2018.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 03/18/2018] [Accepted: 04/16/2018] [Indexed: 02/06/2023]
Abstract
Our previous study demonstrated that obesity aggravated peripheral insulin resistance and brain dysfunction in the ovariectomized condition. Conversely, the effect of obesity followed by oophorectomy on brain oxidative stress, brain apoptosis, synaptic function and cognitive function, particularly in hippocampal-dependent and hippocampal-independent memory, has not been investigated. Our hypothesis was that oophorectomy aggravated metabolic impairment, brain dysfunction and cognitive impairment in obese rats. Thirty-two female rats were fed with either a normal diet (ND, n = 16) or a high-fat diet (HFD, n = 16) for a total of 20 weeks. At week 13, rats in each group were subdivided into sham and ovariectomized subgroups (n = 8/subgroup). At week 20, all rats were tested for hippocampal-dependent and hippocampal-independent memory by using Morris water maze test (MWM) and Novel objective recognition (NOR) tests, respectively. We found that the obese-insulin resistant condition occurred in sham-HFD-fed rats (HFS), ovariectomized-ND-fed rats (NDO), and ovariectomized-HFD-fed rats (HFO). Increased hippocampal oxidative stress level, increased hippocampal apoptosis, increased hippocampal synaptic dysfunction, decreased hippocampal estrogen level and impaired hippocampal-dependent memory were observed in HFS, NDO, and HFO rats. However, the hippocampal-independent memory, cortical estrogen levels, cortical ROS production, and cortical apoptosis showed no significant difference between groups. These findings suggested that oophorectomy and obesity exclusively impaired hippocampal-dependent memory, possibly via increased hippocampal dysfunction. Nonetheless, oophorectomy did not aggravate these deleterious effects under conditions of obesity.
Collapse
Affiliation(s)
- Duangkamol Mantor
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wasana Pratchayasakul
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wanitchaya Minta
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Wissuta Sutham
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siripong Palee
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jirapas Sripetchwandee
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sasiwan Kerdphoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Thidarat Jaiwongkum
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sirawit Sriwichaiin
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Warunsorn Krintratun
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Oral Biology and Diagnostic Science, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
78
|
Grković I, Mitrović N, Dragić M, Adžić M, Drakulić D, Nedeljković N. Spatial Distribution and Expression of Ectonucleotidases in Rat Hippocampus After Removal of Ovaries and Estradiol Replacement. Mol Neurobiol 2018; 56:1933-1945. [PMID: 29978426 DOI: 10.1007/s12035-018-1217-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/29/2018] [Indexed: 01/14/2023]
Abstract
Purinergic signaling is the main synaptic and non-synaptic signaling system in brain. ATP acts as a fast excitatory transmitter, while adenosine sets a global inhibitory tone within hippocampal neuronal networks. ATP and adenosine are interconnected by ectonucleotidase enzymes, which convert ATP to adenosine. Existing data point to the converging roles of ovarian steroids and purinergic signaling in synapse formation and refinement and synapse activity in the hippocampus. Therefore, in the present study, we have used enzyme histochemistry and expression analysis to obtain data on spatial distribution and expression of ecto-enzymes NTPDase1, NTPDase2, and ecto-5'-nucleotidase (eN) after removal of ovaries (OVX) and estradiol replacement (E2) in female rat hippocampus. The results show that target ectonucleotidases are predominantly localized in synapse-rich hippocampal layers. The most represented NTPDase in the hippocampal tissue is NTPDase2, being at the same time the mostly affected ectonucleotidase by OVX and E2. Specifically, OVX decreases the expression of NTPDase2 and eN, whereas E2 restores their expression to control level. Impact of OVX and E2 on ectonucleotidase expression was also examined in purified synaptosome (SYN) and gliosome (GLIO) fractions. Data reveal that SYN expresses NTPDase1 and NTPDase2, both of which are reduced following OVX and restored with E2. GLIO exhibits NTPDase2-mediated ATP hydrolysis, which falls in OVX, and recovers by E2. These changes in the activity occur without parallel changes in NTPDase2-protein abundance. The same holds for eN. The lack of correlation between NTPDase2 and eN activities and their respective protein abundances suggest a non-genomic mode of E2 action, which is studied further in primary astrocyte culture. Since ovarian steroids shape hippocampal synaptic networks and regulate ectonucleotidase activities, it is possible that cognitive deficits seen after ovary removal may arise from the loss of E2 modulatory actions on ectonucleotidase expression in the hippocampus.
Collapse
Affiliation(s)
- Ivana Grković
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, Belgrade, 11001, Serbia.
| | - Nataša Mitrović
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, Belgrade, 11001, Serbia
| | - Milorad Dragić
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, Belgrade, 11001, Serbia
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Studentski trg 3, Belgrade, 11001, Serbia
| | - Marija Adžić
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Studentski trg 3, Belgrade, 11001, Serbia
| | - Dunja Drakulić
- Department of Molecular Biology and Endocrinology, Vinča Institute of Nuclear Sciences, University of Belgrade, Mike Petrovića Alasa 12-14, Belgrade, 11001, Serbia
| | - Nadežda Nedeljković
- Department for General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Studentski trg 3, Belgrade, 11001, Serbia
| |
Collapse
|
79
|
Duarte A, Santos M, Oliveira C, Moreira P. Brain insulin signalling, glucose metabolism and females' reproductive aging: A dangerous triad in Alzheimer's disease. Neuropharmacology 2018; 136:223-242. [PMID: 29471055 DOI: 10.1016/j.neuropharm.2018.01.044] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/22/2018] [Accepted: 01/29/2018] [Indexed: 12/12/2022]
|
80
|
Cloning, partial sequencing and expression analysis of the neural form of P450 aromatase (cyp19a1b) in the South America catfish Rhamdia quelen. Comp Biochem Physiol B Biochem Mol Biol 2018; 221-222:11-17. [DOI: 10.1016/j.cbpb.2018.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/24/2018] [Accepted: 04/02/2018] [Indexed: 02/06/2023]
|
81
|
Denley MCS, Gatford NJF, Sellers KJ, Srivastava DP. Estradiol and the Development of the Cerebral Cortex: An Unexpected Role? Front Neurosci 2018; 12:245. [PMID: 29887794 PMCID: PMC5981095 DOI: 10.3389/fnins.2018.00245] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 03/28/2018] [Indexed: 12/16/2022] Open
Abstract
The cerebral cortex undergoes rapid folding in an "inside-outside" manner during embryonic development resulting in the establishment of six discrete cortical layers. This unique cytoarchitecture occurs via the coordinated processes of neurogenesis and cell migration. In addition, these processes are fine-tuned by a number of extracellular cues, which exert their effects by regulating intracellular signaling pathways. Interestingly, multiple brain regions have been shown to develop in a sexually dimorphic manner. In many cases, estrogens have been demonstrated to play an integral role in mediating these sexual dimorphisms in both males and females. Indeed, 17β-estradiol, the main biologically active estrogen, plays a critical organizational role during early brain development and has been shown to be pivotal in the sexually dimorphic development and regulation of the neural circuitry underlying sex-typical and socio-aggressive behaviors in males and females. However, whether and how estrogens, and 17β-estradiol in particular, regulate the development of the cerebral cortex is less well understood. In this review, we outline the evidence that estrogens are not only present but are engaged and regulate molecular machinery required for the fine-tuning of processes central to the cortex. We discuss how estrogens are thought to regulate the function of key molecular players and signaling pathways involved in corticogenesis, and where possible, highlight if these processes are sexually dimorphic. Collectively, we hope this review highlights the need to consider how estrogens may influence the development of brain regions directly involved in the sex-typical and socio-aggressive behaviors as well as development of sexually dimorphic regions such as the cerebral cortex.
Collapse
Affiliation(s)
- Matthew C. S. Denley
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Nicholas J. F. Gatford
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Katherine J. Sellers
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Deepak P. Srivastava
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| |
Collapse
|
82
|
Del Río JP, Alliende MI, Molina N, Serrano FG, Molina S, Vigil P. Steroid Hormones and Their Action in Women's Brains: The Importance of Hormonal Balance. Front Public Health 2018; 6:141. [PMID: 29876339 PMCID: PMC5974145 DOI: 10.3389/fpubh.2018.00141] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 04/27/2018] [Indexed: 12/19/2022] Open
Abstract
Sex hormones significantly impact women's lives. Throughout the different stages of life, from menarche to menopause and all stages in between, women experience dramatic fluctuations in the levels of progesterone and estradiol, among other hormones. These fluctuations affect the body as a whole, including the central nervous system (CNS). In the CNS, sex hormones act via steroid receptors. They also have an effect on different neurotransmitters such as GABA, serotonin, dopamine, and glutamate. Additionally, studies show that sex hormones and their metabolites influence brain areas that regulate mood, behavior, and cognitive abilities. This review emphasizes the benefits a proper hormonal balance during the different stages of life has in the CNS. To achieve this goal, it is essential that hormone levels are evaluated considering a woman's age and ovulatory status, so that a correct diagnosis and treatment can be made. Knowledge of steroid hormone activity in the brain will give women and health providers an important tool for improving their health and well-being.
Collapse
Affiliation(s)
| | | | | | | | | | - Pilar Vigil
- Reproductive Health Research InstituteSantiago, Chile
- Vicerrectoría de ComunicacionesPontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
83
|
Lejri I, Grimm A, Eckert A. Mitochondria, Estrogen and Female Brain Aging. Front Aging Neurosci 2018; 10:124. [PMID: 29755342 PMCID: PMC5934418 DOI: 10.3389/fnagi.2018.00124] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 04/11/2018] [Indexed: 12/11/2022] Open
Abstract
Mitochondria play an essential role in the generation of steroid hormones including the female sex hormones. These hormones are, in turn, able to modulate mitochondrial activities. Mitochondria possess crucial roles in cell maintenance, survival and well-being, because they are the main source of energy as well as of reactive oxygen species (ROS) within the cell. The impairment of these important organelles is one of the central features of aging. In women’s health, estrogen plays an important role during adulthood not only in the estrous cycle, but also in the brain via neuroprotective, neurotrophic and antioxidant modes of action. The hypestrogenic state in the peri- as well as in the prolonged postmenopause might increase the vulnerability of elderly women to brain degeneration and age-related pathologies. However, the underlying mechanisms that affect these processes are not well elucidated. Understanding the relationship between estrogen and mitochondria might therefore provide better insights into the female aging process. Thus, in this review, we first describe mitochondrial dysfunction in the aging brain. Second, we discuss the estrogen-dependent actions on the mitochondrial activity, including recent evidence of the estrogen—brain-derived neurotrophic factor and estrogen—sirtuin 3 (SIRT3) pathways, as well as their potential implications during female aging.
Collapse
Affiliation(s)
- Imane Lejri
- Neurobiology Lab for Brain Aging and Mental Health, Transfaculty Research Platform Molecular and Cognitive Neuroscience, University of Basel, Basel, Switzerland.,Psychiatric University Clinics, University of Basel, Basel, Switzerland
| | - Amandine Grimm
- Neurobiology Lab for Brain Aging and Mental Health, Transfaculty Research Platform Molecular and Cognitive Neuroscience, University of Basel, Basel, Switzerland.,Psychiatric University Clinics, University of Basel, Basel, Switzerland
| | - Anne Eckert
- Neurobiology Lab for Brain Aging and Mental Health, Transfaculty Research Platform Molecular and Cognitive Neuroscience, University of Basel, Basel, Switzerland.,Psychiatric University Clinics, University of Basel, Basel, Switzerland
| |
Collapse
|
84
|
Lim SW, Nyam TT E, Hu CY, Chio CC, Wang CC, Kuo JR. Estrogen Receptor-α is Involved in Tamoxifen Neuroprotective Effects in a Traumatic Brain Injury Male Rat Model. World Neurosurg 2018; 112:e278-e287. [DOI: 10.1016/j.wneu.2018.01.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 01/06/2018] [Indexed: 01/23/2023]
|
85
|
Alexander BH, Barnes HM, Trimmer E, Davidson AM, Ogola BO, Lindsey SH, Mostany R. Stable Density and Dynamics of Dendritic Spines of Cortical Neurons Across the Estrous Cycle While Expressing Differential Levels of Sensory-Evoked Plasticity. Front Mol Neurosci 2018; 11:83. [PMID: 29615867 PMCID: PMC5864847 DOI: 10.3389/fnmol.2018.00083] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 03/01/2018] [Indexed: 12/11/2022] Open
Abstract
Periodic oscillations of gonadal hormone levels during the estrous cycle exert effects on the female brain, impacting cognition and behavior. While previous research suggests that changes in hormone levels across the cycle affect dendritic spine dynamics in the hippocampus, little is known about the effects on cortical dendritic spines and previous studies showed contradictory results. In this in vivo imaging study, we investigated the impact of the estrous cycle on the density and dynamics of dendritic spines of pyramidal neurons in the primary somatosensory cortex of mice. We also examined if the induction of synaptic plasticity during proestrus, estrus, and metestrus/diestrus had differential effects on the degree of remodeling of synapses in this brain area. We used chronic two-photon excitation (2PE) microscopy during steady-state conditions and after evoking synaptic plasticity by whisker stimulation at the different stages of the cycle. We imaged apical dendritic tufts of layer 5 pyramidal neurons of naturally cycling virgin young female mice. Spine density, turnover rate (TOR), survival fraction, morphology, and volume of mushroom spines remained unaltered across the estrous cycle, and the values of these parameters were comparable with those of young male mice. However, while whisker stimulation of female mice during proestrus and estrus resulted in increases in the TOR of spines (74.2 ± 14.9% and 75.1 ± 12.7% vs. baseline, respectively), sensory-evoked plasticity was significantly lower during metestrus/diestrus (32.3 ± 12.8%). In males, whisker stimulation produced 46.5 ± 20% increase in TOR compared with baseline—not significantly different from female mice at any stage of the cycle. These results indicate that, while steady-state density and dynamics of dendritic spines of layer 5 pyramidal neurons in the primary somatosensory cortex of female mice are constant during the estrous cycle, the susceptibility of these neurons to sensory-evoked structural plasticity may be dependent on the stage of the cycle. Since dendritic spines are more plastic during proestrus and estrus than during metestrus/diestrus, certain stages of the cycle could be more suitable for forms of memory requiring de novo formation and elimination of spines and other stages for forms of memory where retention and/or repurposing of already existing synaptic connections is more pertinent.
Collapse
Affiliation(s)
- Bailin H Alexander
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States
| | - Heather M Barnes
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States.,Neuroscience Program, Brain Institute, Tulane University, New Orleans, LA, United States
| | - Emma Trimmer
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States
| | - Andrew M Davidson
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States.,Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, United States
| | - Benard O Ogola
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States.,Brain Institute, Tulane University, New Orleans, LA, United States
| | - Ricardo Mostany
- Department of Pharmacology, Tulane University School of Medicine, Tulane University, New Orleans, LA, United States.,Brain Institute, Tulane University, New Orleans, LA, United States
| |
Collapse
|
86
|
Marin R, Diaz M. Estrogen Interactions With Lipid Rafts Related to Neuroprotection. Impact of Brain Ageing and Menopause. Front Neurosci 2018; 12:128. [PMID: 29559883 PMCID: PMC5845729 DOI: 10.3389/fnins.2018.00128] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 02/16/2018] [Indexed: 12/22/2022] Open
Abstract
Estrogens (E2) exert a plethora of neuroprotective actions against aged-associated brain diseases, including Alzheimer's disease (AD). Part of these actions takes place through binding to estrogen receptors (ER) embedded in signalosomes, where numerous signaling proteins are clustered. Signalosomes are preferentially located in lipid rafts which are dynamic membrane microstructures characterized by a peculiar lipid composition enriched in gangliosides, saturated fatty acids, cholesterol, and sphingolipids. Rapid E2 interactions with ER-related signalosomes appear to trigger intracellular signaling ultimately leading to the activation of molecular mechanisms against AD. We have previously observed that the reduction of E2 blood levels occurring during menopause induced disruption of ER-signalosomes at frontal cortical brain areas. These molecular changes may reduce neuronal protection activities, as similar ER signalosome derangements were observed in AD brains. The molecular impairments may be associated with changes in the lipid composition of lipid rafts observed in neurons during menopause and AD. These evidences indicate that the changes in lipid raft structure during aging may be at the basis of alterations in the activity of ER and other neuroprotective proteins integrated in these membrane microstructures. Moreover, E2 is a homeostatic modulator of lipid rafts. Recent work has pointed to this relevant aspect of E2 activity to preserve brain integrity, through mechanisms affecting lipid uptake and local biosynthesis in the brain. Some evidences have demonstrated that estrogens and the docosahexaenoic acid (DHA) exert synergistic effects to stabilize brain lipid matrix. DHA is essential to enhance molecular fluidity at the plasma membrane, promoting functional macromolecular interactions in signaling platforms. In support of this, DHA detriment in neuronal lipid rafts has been associated with the most common age-associated neuropathologies, namely AD and Parkinson disease. Altogether, these findings indicate that E2 may participate in brain preservation through a dual membrane-related mechanism. On the one hand, E2 interacting with ER related signalosomes may protect against neurotoxic insults. On the other hand, E2 may exert lipostatic actions to preserve lipid balance in neuronal membrane microdomains. The different aspects of the emerging multifunctional role of estrogens in membrane-related signalosomes will be discussed in this review.
Collapse
Affiliation(s)
- Raquel Marin
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Medicine, Faculty of Health Sciences, University of La Laguna, Tenerife, Spain.,Fisiología y Biofísica de la Membrana Celular en Patologías Neurodegenerativas y Tumorales, Consejo Superior de Investigaciones Cientificas, Unidad Asociada de Investigación, Universidad de La Laguna Tenerife, Tenerife, Spain
| | - Mario Diaz
- Fisiología y Biofísica de la Membrana Celular en Patologías Neurodegenerativas y Tumorales, Consejo Superior de Investigaciones Cientificas, Unidad Asociada de Investigación, Universidad de La Laguna Tenerife, Tenerife, Spain.,Laboratory of Membrane Physiology and Biophysics, Department of Animal Biology, Edaphology and Geology, University of La Laguna, Tenerife, Spain
| |
Collapse
|
87
|
Jacobs EG, Weiss B, Makris N, Whitfield-Gabrieli S, Buka SL, Klibanski A, Goldstein JM. Reorganization of Functional Networks in Verbal Working Memory Circuitry in Early Midlife: The Impact of Sex and Menopausal Status. Cereb Cortex 2018; 27:2857-2870. [PMID: 27178194 DOI: 10.1093/cercor/bhw127] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Converging preclinical and human evidence indicates that the decline in ovarian estradiol production during the menopausal transition may play a mechanistic role in the neuronal changes that occur early in the aging process. Here, we present findings from a population-based fMRI study characterizing regional and network-level differences in working memory (WM) circuitry in midlife men and women (N = 142; age range 46-53), as a function of sex and reproductive stage. Reproductive histories and hormonal evaluations were used to determine menopausal status. Participants performed a verbal WM task during fMRI scanning. Results revealed robust differences in task-evoked responses in dorsolateral prefrontal cortex and hippocampus as a function of women's reproductive stage, despite minimal variance in chronological age. Sex differences in regional activity and functional connectivity that were pronounced between men and premenopausal women were diminished for postmenopausal women. Critically, analyzing data without regard to sex or reproductive status obscured group differences in the circuit-level neural strategies associated with successful working memory performance. These findings underscore the importance of reproductive age and hormonal status, over and above chronological age, for understanding sex differences in the aging of memory circuitry. Further, these findings suggest that early changes in working memory circuitry are evident decades before the age range typically targeted in cognitive aging studies.
Collapse
Affiliation(s)
- Emily G Jacobs
- Division of Women's Health, Department of Medicine.,Department of Psychiatry, Brigham and Women's Hospital, Boston, MA 02120, USA.,Harvard Medical School, Boston, MA 02120, USA.,Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Blair Weiss
- Department of Psychiatry, Brigham and Women's Hospital, Boston, MA 02120, USA
| | - Nikos Makris
- Harvard Medical School, Boston, MA 02120, USA.,Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | - Sue Whitfield-Gabrieli
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Stephen L Buka
- Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA
| | - Anne Klibanski
- Neuroendocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jill M Goldstein
- Division of Women's Health, Department of Medicine.,Department of Psychiatry, Brigham and Women's Hospital, Boston, MA 02120, USA.,Harvard Medical School, Boston, MA 02120, USA.,Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| |
Collapse
|
88
|
Abstract
OBJECTIVE Few have characterized cognitive changes with age as a function of menopausal stage relative to men, or sex differences in components of memory in early midlife. The study aim was to investigate variation in memory function in early midlife as a function of sex, sex steroid hormones, and reproductive status. METHODS A total of 212 men and women aged 45 to 55 were selected for this cross-sectional study from a prenatal cohort of pregnancies whose mothers were originally recruited in 1959 to 1966. They underwent clinical and cognitive testing and hormonal assessments of menopause status. Multivariate general linear models for multiple memory outcomes were used to test hypotheses controlling for potential confounders. Episodic memory, executive function, semantic processing, and estimated verbal intelligence were assessed. Associative memory and episodic verbal memory were assessed using Face-Name Associative Memory Exam (FNAME) and Selective Reminding Test (SRT), given increased sensitivity to detecting early cognitive decline. Impacts of sex and reproductive stage on performance were tested. RESULTS Women outperformed men on all memory measures including FNAME (β = -0.30, P < 0.0001) and SRT (β = -0.29, P < 0.0001). Furthermore, premenopausal and perimenopausal women outperformed postmenopausal women on FNAME (initial learning, β= 0.32, P = 0.01) and SRT (recall, β= 2.39, P = 0.02). Across all women, higher estradiol was associated with better SRT performance (recall, β = 1.96, P = 0.01) and marginally associated with FNAME (initial learning, β = 0.19, P = 0.06). CONCLUSIONS This study demonstrated that, in early midlife, women outperformed age-matched men across all memory measures, but sex differences were attenuated for postmenopausal women. Initial learning and memory retrieval were particularly vulnerable, whereas memory consolidation and storage were preserved. Findings underscore the significance of the decline in ovarian estradiol production in midlife and its role in shaping memory function.
Collapse
|
89
|
Frozza RL, Lourenco MV, De Felice FG. Challenges for Alzheimer's Disease Therapy: Insights from Novel Mechanisms Beyond Memory Defects. Front Neurosci 2018; 12:37. [PMID: 29467605 PMCID: PMC5808215 DOI: 10.3389/fnins.2018.00037] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/16/2018] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease (AD), the most common form of dementia in late life, will become even more prevalent by midcentury, constituting a major global health concern with huge implications for individuals and society. Despite scientific breakthroughs during the past decades that have expanded our knowledge on the cellular and molecular bases of AD, therapies that effectively halt disease progression are still lacking, and focused efforts are needed to address this public health challenge. Because AD is classically recognized as a disease of memory, studies have mainly focused on investigating memory-associated brain defects. However, compelling evidence has indicated that additional brain regions, not classically linked to memory, are also affected in the course of disease. In this review, we outline the current understanding of key pathophysiological mechanisms in AD and their clinical manifestation. We also highlight how considering the complex nature of AD pathogenesis, and exploring repurposed drug approaches can pave the road toward the development of novel therapeutics for AD.
Collapse
Affiliation(s)
- Rudimar L. Frozza
- Oswaldo Cruz Institute, Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Mychael V. Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Rio de Janeiro, Brazil
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernanda G. De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Rio de Janeiro, Brazil
- Department of Biomedical and Molecular Sciences, Centre for Neuroscience Studies, Queen's University, Kingston, ON, Canada
| |
Collapse
|
90
|
Kantarci K, Lowe VJ, Lesnick TG, Tosakulwong N, Bailey KR, Fields JA, Shuster LT, Zuk SM, Senjem ML, Mielke MM, Gleason C, Jack CR, Rocca WA, Miller VM. Early Postmenopausal Transdermal 17β-Estradiol Therapy and Amyloid-β Deposition. J Alzheimers Dis 2018; 53:547-56. [PMID: 27163830 PMCID: PMC4955514 DOI: 10.3233/jad-160258] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Background: It remains controversial whether hormone therapy in recently postmenopausal women modifies the risk of Alzheimer’s disease (AD). Objective: To investigate the effects of hormone therapy on amyloid-β deposition in recently postmenopausal women. Methods: Participants within 5–36 months past menopause in the Kronos Early Estrogen Prevention Study, a randomized, double blinded placebo-controlled clinical trial, were randomized to: 1) 0.45 mg/day oral conjugated equine estrogens (CEE); 2) 50μg/day transdermal 17β-estradiol; or 3) placebo pills and patch for four years. Oral progesterone (200 mg/day) was given to active treatment groups for 12 days each month. 11C Pittsburgh compound B (PiB) PET imaging was performed in 68 of the 118 participants at Mayo Clinic approximately seven years post randomization and three years after stopping randomized treatment. PiB Standard unit value ratio (SUVR) was calculated. Results: Women (age = 52–65) randomized to transdermal 17β-estradiol (n = 21) had lower PiB SUVR compared to placebo (n = 30) after adjusting for age [odds ratio (95% CI) = 0.31(0.11–0.83)]. In the APOEɛ4 carriers, transdermal 17β-estradiol treated women (n = 10) had lower PiB SUVR compared to either placebo (n = 5) [odds ratio (95% CI) = 0.04(0.004–0.44)], or the oral CEE treated group (n = 3) [odds ratio (95% CI) = 0.01(0.0006–0.23)] after adjusting for age. Hormone therapy was not associated with PiB SUVR in the APOEɛ4 non-carriers. Conclusion: In this pilot study, transdermal 17β-estradiol therapy in recently postmenopausal women was associated with a reduced amyloid-β deposition, particularly in APOEɛ4 carriers. This finding may have important implications for the prevention of AD in postmenopausal women, and needs to be confirmed in a larger sample.
Collapse
Affiliation(s)
- Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Timothy G Lesnick
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | | | - Kent R Bailey
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Julie A Fields
- Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA
| | - Lynne T Shuster
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Samantha M Zuk
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Matthew L Senjem
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | - Michelle M Mielke
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA.,Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Carey Gleason
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin and Geriatric Research, Education and Clinical Center (GRECC) William S. Middleton Memorial, Veterans' Hospital, Madison, WI, USA
| | | | - Walter A Rocca
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA.,Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Virginia M Miller
- Departments of Surgery, Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
91
|
Zárate S, Stevnsner T, Gredilla R. Role of Estrogen and Other Sex Hormones in Brain Aging. Neuroprotection and DNA Repair. Front Aging Neurosci 2018. [PMID: 29311911 DOI: 10.3389/fnagi.2017.00430/xml/nlm] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Aging is an inevitable biological process characterized by a progressive decline in physiological function and increased susceptibility to disease. The detrimental effects of aging are observed in all tissues, the brain being the most important one due to its main role in the homeostasis of the organism. As our knowledge about the underlying mechanisms of brain aging increases, potential approaches to preserve brain function rise significantly. Accumulating evidence suggests that loss of genomic maintenance may contribute to aging, especially in the central nervous system (CNS) owing to its low DNA repair capacity. Sex hormones, particularly estrogens, possess potent antioxidant properties and play important roles in maintaining normal reproductive and non-reproductive functions. They exert neuroprotective actions and their loss during aging and natural or surgical menopause is associated with mitochondrial dysfunction, neuroinflammation, synaptic decline, cognitive impairment and increased risk of age-related disorders. Moreover, loss of sex hormones has been suggested to promote an accelerated aging phenotype eventually leading to the development of brain hypometabolism, a feature often observed in menopausal women and prodromal Alzheimer's disease (AD). Although data on the relation between sex hormones and DNA repair mechanisms in the brain is still limited, various investigations have linked sex hormone levels with different DNA repair enzymes. Here, we review estrogen anti-aging and neuroprotective mechanisms, which are currently an area of intense study, together with the effect they may have on the DNA repair capacity in the brain.
Collapse
Affiliation(s)
- Sandra Zárate
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Tinna Stevnsner
- Danish Center for Molecular Gerontology and Danish Aging Research Center, Department of Molecular Biology and Genetics, University of Aarhus, Aarhus, Denmark
| | - Ricardo Gredilla
- Department of Physiology, Faculty of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
92
|
Scheyer O, Rahman A, Hristov H, Berkowitz C, Isaacson RS, Diaz Brinton R, Mosconi L. Female Sex and Alzheimer's Risk: The Menopause Connection. J Prev Alzheimers Dis 2018; 5:225-230. [PMID: 30298180 PMCID: PMC6198681 DOI: 10.14283/jpad.2018.34] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Along with advanced age and apolipoprotein E (APOE)-4 genotype, female sex is a major risk factor for developing late-onset Alzheimer's disease (AD). Considering that AD pathology begins decades prior to clinical symptoms, the higher risk in women cannot simply be accounted for by their greater longevity as compared to men. Recent investigation into sex-specific pathophysiological mechanisms behind AD risk has implicated the menopause transition (MT), a midlife neuroendocrine transition state unique to females. Commonly characterized as ending in reproductive senescence, many symptoms of MT are neurological, including disruption of estrogen-regulated systems such as thermoregulation, sleep, and circadian rhythms, as well as depression and impairment in multiple cognitive domains. Preclinical studies have shown that, during MT, the estrogen network uncouples from the brain bioenergetic system. The resulting hypometabolic state could serve as the substrate for neurological dysfunction. Indeed, translational brain imaging studies demonstrate that 40-60 year-old perimenopausal and postmenopausal women exhibit an AD-endophenotype characterized by decreased metabolic activity and increased brain amyloid-beta deposition as compared to premenopausal women and to age-matched men. This review discusses the MT as a window of opportunity for therapeutic interventions to compensate for brain bioenergetic crisis and combat the subsequent increased risk for AD in women.
Collapse
Affiliation(s)
- O Scheyer
- Lisa Mosconi, PhD, Department of Neurology, Weill Cornell Medicine, 428 East 72nd St, Suite 500, Room 407, New York, NY, 10021; Tel: (212) 746-4624,
| | | | | | | | | | | | | |
Collapse
|
93
|
Berger J, Demin K, Holtkamp M, Bengner T. Female verbal memory advantage in temporal, but not frontal lobe epilepsy. Epilepsy Res 2018; 139:129-134. [DOI: 10.1016/j.eplepsyres.2017.11.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/22/2017] [Accepted: 11/28/2017] [Indexed: 02/07/2023]
|
94
|
Zárate S, Stevnsner T, Gredilla R. Role of Estrogen and Other Sex Hormones in Brain Aging. Neuroprotection and DNA Repair. Front Aging Neurosci 2017; 9:430. [PMID: 29311911 PMCID: PMC5743731 DOI: 10.3389/fnagi.2017.00430] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 12/14/2017] [Indexed: 12/13/2022] Open
Abstract
Aging is an inevitable biological process characterized by a progressive decline in physiological function and increased susceptibility to disease. The detrimental effects of aging are observed in all tissues, the brain being the most important one due to its main role in the homeostasis of the organism. As our knowledge about the underlying mechanisms of brain aging increases, potential approaches to preserve brain function rise significantly. Accumulating evidence suggests that loss of genomic maintenance may contribute to aging, especially in the central nervous system (CNS) owing to its low DNA repair capacity. Sex hormones, particularly estrogens, possess potent antioxidant properties and play important roles in maintaining normal reproductive and non-reproductive functions. They exert neuroprotective actions and their loss during aging and natural or surgical menopause is associated with mitochondrial dysfunction, neuroinflammation, synaptic decline, cognitive impairment and increased risk of age-related disorders. Moreover, loss of sex hormones has been suggested to promote an accelerated aging phenotype eventually leading to the development of brain hypometabolism, a feature often observed in menopausal women and prodromal Alzheimer's disease (AD). Although data on the relation between sex hormones and DNA repair mechanisms in the brain is still limited, various investigations have linked sex hormone levels with different DNA repair enzymes. Here, we review estrogen anti-aging and neuroprotective mechanisms, which are currently an area of intense study, together with the effect they may have on the DNA repair capacity in the brain.
Collapse
Affiliation(s)
- Sandra Zárate
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Histología, Embriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Tinna Stevnsner
- Danish Center for Molecular Gerontology and Danish Aging Research Center, Department of Molecular Biology and Genetics, University of Aarhus, Aarhus, Denmark
| | - Ricardo Gredilla
- Department of Physiology, Faculty of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
95
|
Disruption of Interneuron Neurogenesis in Premature Newborns and Reversal with Estrogen Treatment. J Neurosci 2017; 38:1100-1113. [PMID: 29246927 DOI: 10.1523/jneurosci.1875-17.2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/19/2017] [Accepted: 10/26/2017] [Indexed: 11/21/2022] Open
Abstract
Many Preterm-born children suffer from neurobehavioral disorders. Premature birth terminates the hypoxic in utero environment and supply of maternal hormones. As the production of interneurons continues until the end of pregnancy, we hypothesized that premature birth would disrupt interneuron production and that restoration of the hypoxic milieu or estrogen treatment might reverse interneuron generation. To test these hypotheses, we compared interneuronal progenitors in the medial ganglionic eminences (MGEs), lateral ganglionic eminences (LGEs), and caudal ganglionic eminences (CGEs) between preterm-born [born on embryonic day (E) 29; examined on postnatal day (D) 3 and D7] and term-born (born on E32; examined on D0 and D4) rabbits at equivalent postconceptional ages. We found that both total and cycling Nkx2.1+, Dlx2+, and Sox2+ cells were more abundant in the MGEs of preterm rabbits at D3 compared with term rabbits at D0, but not in D7 preterm relative to D4 term pups. Total Nkx2.1+ progenitors were also more numerous in the LGEs of preterm pups at D3 compared with term rabbits at D0. Dlx2+ cells in CGEs were comparable between preterm and term pups. Simulation of hypoxia by dimethyloxalylglycine treatment did not affect the number of interneuronal progenitors. However, estrogen treatment reduced the density of total and proliferating Nkx2.1+ and Dlx2+ cells in the MGEs and enhanced Ascl1 transcription factor. Estrogen treatment also reduced Ki67, c-Myc, and phosphorylation of retinoblastoma protein, suggesting inhibition of the G1-to-S phase transition. Hence, preterm birth disrupts interneuron neurogenesis in the MGE and estrogen treatment reverses interneuron neurogenesis in preterm newborns by cell-cycle inhibition and elevation of Ascl1. We speculate that estrogen replacement might partially restore neurogenesis in human premature infants.SIGNIFICANCE STATEMENT Prematurity results in developmental delays and neurobehavioral disorders, which might be ascribed to disturbances in the development of cortical interneurons. Here, we show that preterm birth disrupts interneuron neurogenesis in the medial ganglionic eminence (MGE) and, more importantly, that estrogen treatment reverses this perturbation in the population of interneuron progenitors in the MGE. The estrogen seems to restore neurogenesis by inhibiting the cell cycle and elevating Ascl1 expression. As preterm birth causes plasma estrogen level to drop 100-fold, the estrogen replacement in preterm infants is physiological. We speculate that estrogen replacement might ameliorate disruption in production of interneurons in human premature infants.
Collapse
|
96
|
Pike CJ. Sex and the development of Alzheimer's disease. J Neurosci Res 2017; 95:671-680. [PMID: 27870425 DOI: 10.1002/jnr.23827] [Citation(s) in RCA: 285] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/17/2016] [Accepted: 06/20/2016] [Indexed: 12/20/2022]
Abstract
Men and women exhibit differences in the development and progression of Alzheimer's disease (AD). The factors underlying the sex differences in AD are not well understood. This Review emphasizes the contributions of sex steroid hormones to the relationship between sex and AD. In women, events that decrease lifetime exposure to estrogens are generally associated with increased AD risk, whereas estrogen-based hormone therapy administered near the time of menopause may reduce AD risk. In men, estrogens do not exhibit age-related reduction and are not significantly associated with AD risk. Rather, normal age-related depletions of testosterone in plasma and brain predict enhanced vulnerability to AD. Both estrogens and androgens exert numerous protective actions in the adult brain that increase neural functioning and resilience as well as specifically attenuating multiple aspects of AD-related neuropathology. Aging diminishes the activational effects of sex hormones in sex-specific manners, which is hypothesized to contribute to the relationship between aging and AD. Sex steroid hormones may also drive sex differences in AD through their organizational effects during developmental sexual differentiation of the brain. Specifically, sex hormone actions during early development may confer inherent vulnerability of the female brain to development of AD in advanced age. The combined effects of organizational and activational effects of sex steroids yield distinct sex differences in AD pathogenesis, a significant variable that must be more rigorously considered in future research. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Christian J Pike
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California
| |
Collapse
|
97
|
The effect of sex and menstrual phase on memory formation during a nap. Neurobiol Learn Mem 2017; 145:119-128. [DOI: 10.1016/j.nlm.2017.09.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/08/2017] [Accepted: 09/12/2017] [Indexed: 01/20/2023]
|
98
|
Bao J, Mahaman YAR, Liu R, Wang JZ, Zhang Z, Zhang B, Wang X. Sex Differences in the Cognitive and Hippocampal Effects of Streptozotocin in an Animal Model of Sporadic AD. Front Aging Neurosci 2017; 9:347. [PMID: 29163130 PMCID: PMC5671606 DOI: 10.3389/fnagi.2017.00347] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 10/16/2017] [Indexed: 01/07/2023] Open
Abstract
More than 95% of Alzheimer's disease (AD) belongs to sporadic AD (sAD), and related animal models are the important research tools for investigating the pathogenesis and developing new drugs for sAD. An intracerebroventricular infusion of streptozotocin (ICV-STZ) is commonly employed to generate sporadic AD animal model. Moreover, the potential impact of sex on brain function is now emphasized in the field of AD. However, whether sex differences exist in AD animal models remains unknown. Here we reported that ICV-STZ remarkably resulted in learning and memory impairment in the Sprague-Dawley male rats, but not in the female rats. We also found tau hyperphosphorylation, an increase of Aβ40/42 as well as increase in both GSK-3β and BACE1 activities, while a loss of dendritic and synaptic plasticity was observed in the male STZ rats. However, STZ did not induce above alterations in the female rats. Furthermore, estradiol levels of serum and hippocampus of female rats were much higher than that of male rats. In conclusion, sex differences exist in this sporadic AD animal model (Sprague-Dawley rats induced by STZ), and this should be considered in future AD research.
Collapse
Affiliation(s)
- Jian Bao
- Key Laboratory of Ministry of Education of China for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yacoubou A R Mahaman
- Key Laboratory of Ministry of Education of China for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Liu
- Key Laboratory of Ministry of Education of China for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- Key Laboratory of Ministry of Education of China for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Zhiguo Zhang
- School of Medicine and Health Management, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Xiaochuan Wang
- Key Laboratory of Ministry of Education of China for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
99
|
Mosconi L, Berti V, Quinn C, McHugh P, Petrongolo G, Osorio RS, Connaughty C, Pupi A, Vallabhajosula S, Isaacson RS, de Leon MJ, Swerdlow RH, Brinton RD. Perimenopause and emergence of an Alzheimer's bioenergetic phenotype in brain and periphery. PLoS One 2017; 12:e0185926. [PMID: 29016679 PMCID: PMC5634623 DOI: 10.1371/journal.pone.0185926] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 09/21/2017] [Indexed: 01/07/2023] Open
Abstract
After advanced age, female sex is the major risk factor for Alzheimer’s disease (AD). The biological mechanisms underlying the increased AD risk in women remain largely undetermined. Preclinical studies identified the perimenopause to menopause transition, a neuroendocrine transition state unique to the female, as a sex-specific risk factor for AD. In animals, estrogenic regulation of cerebral glucose metabolism (CMRglc) falters during perimenopause. This is evident in glucose hypometabolism and decline in mitochondrial efficiency which is sustained thereafter. This study bridges basic to clinical science to characterize brain bioenergetics in a cohort of forty-three, 40–60 year-old clinically and cognitively normal women at different endocrine transition stages including premenopause (controls, CNT, n = 15), perimenopause (PERI, n = 14) and postmenopause (MENO, n = 14). All participants received clinical, laboratory and neuropsychological examinations, 18F-fluoro-deoxyglucose (FDG)-Positron Emission Tomography (PET) FDG-PET scans to estimate CMRglc, and platelet mitochondrial cytochrome oxidase (COX) activity measures. Statistical parametric mapping and multiple regression models were used to examine clinical, CMRglc and COX data across groups. As expected, the MENO group was older than PERI and controls. Groups were otherwise comparable for clinical measures and distribution of APOE4 genotype. Both MENO and PERI groups exhibited reduced CMRglc in AD-vulnerable regions which was correlated with decline in mitochondrial COX activity compared to CNT (p’s<0.001). A gradient in biomarker abnormalities was most pronounced in MENO, intermediate in PERI, and lowest in CNT (p<0.001). Biomarkers correlated with immediate and delayed memory scores (Pearson’s 0.26≤r≤0.32, p≤0.05). These findings validate earlier preclinical findings and indicate emergence of bioenergetic deficits in perimenopausal and postmenopausal women, suggesting that the optimal window of opportunity for therapeutic intervention in women is early in the endocrine aging process.
Collapse
Affiliation(s)
- Lisa Mosconi
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States of America.,Department of Psychiatry, New York University School of Medicine, New York, NY, United States of America
| | - Valentina Berti
- Department of Clinical Pathophysiology, Nuclear Medicine Unit, University of Florence, Florence, Italy
| | - Crystal Quinn
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States of America
| | - Pauline McHugh
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States of America
| | - Gabriella Petrongolo
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States of America
| | - Ricardo S Osorio
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States of America
| | - Christopher Connaughty
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States of America
| | - Alberto Pupi
- Department of Clinical Pathophysiology, Nuclear Medicine Unit, University of Florence, Florence, Italy
| | - Shankar Vallabhajosula
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States of America
| | - Richard S Isaacson
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States of America
| | - Mony J de Leon
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States of America
| | - Russell H Swerdlow
- Department of Neurology, University of Kansas School of Medicine, Kansas City, United States of America
| | - Roberta Diaz Brinton
- Departments of Pharmacology and Neurology, University of Arizona College of Medicine, Tucson, AZ, United States of America
| |
Collapse
|
100
|
Mosconi L, Berti V, Quinn C, McHugh P, Petrongolo G, Varsavsky I, Osorio RS, Pupi A, Vallabhajosula S, Isaacson RS, de Leon MJ, Brinton RD. Sex differences in Alzheimer risk: Brain imaging of endocrine vs chronologic aging. Neurology 2017; 89:1382-1390. [PMID: 28855400 DOI: 10.1212/wnl.0000000000004425] [Citation(s) in RCA: 210] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 07/05/2017] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE This observational multimodality brain imaging study investigates emergence of endophenotypes of late-onset Alzheimer disease (AD) risk during endocrine transition states in a cohort of clinically and cognitively normal women and age-matched men. METHODS Forty-two 40- to 60-year-old cognitively normal women (15 asymptomatic perimenopausal by age [CNT], 13 perimenopausal [PERI], and 14 postmenopausal [MENO]) and 18 age- and education-matched men were examined. All patients had volumetric MRI, 18F-fluoro-2-deoxyglucose (FDG)-PET (glucose metabolism), and Pittsburgh compound B-PET scans (β-amyloid [Aβ] deposition, a hallmark of AD pathology). RESULTS As expected, the MENO group was older than the PERI and CNT groups. Otherwise, groups were comparable on clinical and neuropsychological measures and APOE4 distribution. Compared to CNT women and to men, and controlling for age, PERI and MENO groups exhibited increased indicators of AD endophenotype, including hypometabolism, increased Aβ deposition, and reduced gray and white matter volumes in AD-vulnerable regions (p < 0.001). AD biomarker abnormalities were greatest in MENO, intermediate in PERI, and lowest in CNT women (p < 0.001). Aβ deposition was exacerbated in APOE4-positive MENO women relative to the other groups (p < 0.001). CONCLUSIONS Multimodality brain imaging indicates sex differences in development of the AD endophenotype, suggesting that the preclinical AD phase is early in the female aging process and coincides with the endocrine transition of perimenopause. These data indicate that the optimal window of opportunity for therapeutic intervention in women is early in the endocrine aging process.
Collapse
Affiliation(s)
- Lisa Mosconi
- From the Departments of Neurology (L.M., R.S.I.) and Radiology (S.V.), Weill Cornell Medical College; Department of Psychiatry (L.M., C.Q., P.M., G.P., I.V., R.S.O., M.J.d.L.), New York University School of Medicine, New York; Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Departments of Pharmacology and Neurology (R.D.B.), College of Medicine, University of Arizona, Tucson; and Departments of Pharmacology, Biomedical Engineering, and Neurology (R.D.B.), University of South California, Los Angeles.
| | - Valentina Berti
- From the Departments of Neurology (L.M., R.S.I.) and Radiology (S.V.), Weill Cornell Medical College; Department of Psychiatry (L.M., C.Q., P.M., G.P., I.V., R.S.O., M.J.d.L.), New York University School of Medicine, New York; Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Departments of Pharmacology and Neurology (R.D.B.), College of Medicine, University of Arizona, Tucson; and Departments of Pharmacology, Biomedical Engineering, and Neurology (R.D.B.), University of South California, Los Angeles
| | - Crystal Quinn
- From the Departments of Neurology (L.M., R.S.I.) and Radiology (S.V.), Weill Cornell Medical College; Department of Psychiatry (L.M., C.Q., P.M., G.P., I.V., R.S.O., M.J.d.L.), New York University School of Medicine, New York; Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Departments of Pharmacology and Neurology (R.D.B.), College of Medicine, University of Arizona, Tucson; and Departments of Pharmacology, Biomedical Engineering, and Neurology (R.D.B.), University of South California, Los Angeles
| | - Pauline McHugh
- From the Departments of Neurology (L.M., R.S.I.) and Radiology (S.V.), Weill Cornell Medical College; Department of Psychiatry (L.M., C.Q., P.M., G.P., I.V., R.S.O., M.J.d.L.), New York University School of Medicine, New York; Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Departments of Pharmacology and Neurology (R.D.B.), College of Medicine, University of Arizona, Tucson; and Departments of Pharmacology, Biomedical Engineering, and Neurology (R.D.B.), University of South California, Los Angeles
| | - Gabriella Petrongolo
- From the Departments of Neurology (L.M., R.S.I.) and Radiology (S.V.), Weill Cornell Medical College; Department of Psychiatry (L.M., C.Q., P.M., G.P., I.V., R.S.O., M.J.d.L.), New York University School of Medicine, New York; Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Departments of Pharmacology and Neurology (R.D.B.), College of Medicine, University of Arizona, Tucson; and Departments of Pharmacology, Biomedical Engineering, and Neurology (R.D.B.), University of South California, Los Angeles
| | - Isabella Varsavsky
- From the Departments of Neurology (L.M., R.S.I.) and Radiology (S.V.), Weill Cornell Medical College; Department of Psychiatry (L.M., C.Q., P.M., G.P., I.V., R.S.O., M.J.d.L.), New York University School of Medicine, New York; Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Departments of Pharmacology and Neurology (R.D.B.), College of Medicine, University of Arizona, Tucson; and Departments of Pharmacology, Biomedical Engineering, and Neurology (R.D.B.), University of South California, Los Angeles
| | - Ricardo S Osorio
- From the Departments of Neurology (L.M., R.S.I.) and Radiology (S.V.), Weill Cornell Medical College; Department of Psychiatry (L.M., C.Q., P.M., G.P., I.V., R.S.O., M.J.d.L.), New York University School of Medicine, New York; Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Departments of Pharmacology and Neurology (R.D.B.), College of Medicine, University of Arizona, Tucson; and Departments of Pharmacology, Biomedical Engineering, and Neurology (R.D.B.), University of South California, Los Angeles
| | - Alberto Pupi
- From the Departments of Neurology (L.M., R.S.I.) and Radiology (S.V.), Weill Cornell Medical College; Department of Psychiatry (L.M., C.Q., P.M., G.P., I.V., R.S.O., M.J.d.L.), New York University School of Medicine, New York; Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Departments of Pharmacology and Neurology (R.D.B.), College of Medicine, University of Arizona, Tucson; and Departments of Pharmacology, Biomedical Engineering, and Neurology (R.D.B.), University of South California, Los Angeles
| | - Shankar Vallabhajosula
- From the Departments of Neurology (L.M., R.S.I.) and Radiology (S.V.), Weill Cornell Medical College; Department of Psychiatry (L.M., C.Q., P.M., G.P., I.V., R.S.O., M.J.d.L.), New York University School of Medicine, New York; Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Departments of Pharmacology and Neurology (R.D.B.), College of Medicine, University of Arizona, Tucson; and Departments of Pharmacology, Biomedical Engineering, and Neurology (R.D.B.), University of South California, Los Angeles
| | - Richard S Isaacson
- From the Departments of Neurology (L.M., R.S.I.) and Radiology (S.V.), Weill Cornell Medical College; Department of Psychiatry (L.M., C.Q., P.M., G.P., I.V., R.S.O., M.J.d.L.), New York University School of Medicine, New York; Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Departments of Pharmacology and Neurology (R.D.B.), College of Medicine, University of Arizona, Tucson; and Departments of Pharmacology, Biomedical Engineering, and Neurology (R.D.B.), University of South California, Los Angeles
| | - Mony J de Leon
- From the Departments of Neurology (L.M., R.S.I.) and Radiology (S.V.), Weill Cornell Medical College; Department of Psychiatry (L.M., C.Q., P.M., G.P., I.V., R.S.O., M.J.d.L.), New York University School of Medicine, New York; Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Departments of Pharmacology and Neurology (R.D.B.), College of Medicine, University of Arizona, Tucson; and Departments of Pharmacology, Biomedical Engineering, and Neurology (R.D.B.), University of South California, Los Angeles
| | - Roberta Diaz Brinton
- From the Departments of Neurology (L.M., R.S.I.) and Radiology (S.V.), Weill Cornell Medical College; Department of Psychiatry (L.M., C.Q., P.M., G.P., I.V., R.S.O., M.J.d.L.), New York University School of Medicine, New York; Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (V.B., A.P.), Nuclear Medicine Unit, University of Florence, Italy; Departments of Pharmacology and Neurology (R.D.B.), College of Medicine, University of Arizona, Tucson; and Departments of Pharmacology, Biomedical Engineering, and Neurology (R.D.B.), University of South California, Los Angeles
| |
Collapse
|