1
|
Shao X, Shou Q, Felix K, Ojogho B, Jiang X, Gold BT, Herting MM, Goldwaser EL, Kochunov P, Hong E, Pappas I, Braskie M, Kim H, Cen S, Jann K, Wang DJJ. Age-related decline in blood-brain barrier function is more pronounced in males than females in parietal and temporal regions. eLife 2024; 13:RP96155. [PMID: 39495221 PMCID: PMC11534331 DOI: 10.7554/elife.96155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024] Open
Abstract
The blood-brain barrier (BBB) plays a pivotal role in protecting the central nervous system (CNS), and shielding it from potential harmful entities. A natural decline of BBB function with aging has been reported in both animal and human studies, which may contribute to cognitive decline and neurodegenerative disorders. Limited data also suggest that being female may be associated with protective effects on BBB function. Here, we investigated age and sex-dependent trajectories of perfusion and BBB water exchange rate (kw) across the lifespan in 186 cognitively normal participants spanning the ages of 8-92 years old, using a non-invasive diffusion-prepared pseudo-continuous arterial spin labeling (DP-pCASL) MRI technique. We found that the pattern of BBB kw decline with aging varies across brain regions. Moreover, results from our DP-pCASL technique revealed a remarkable decline in BBB kw beginning in the early 60 s, which was more pronounced in males. In addition, we observed sex differences in parietal and temporal regions. Our findings provide in vivo results demonstrating sex differences in the decline of BBB function with aging, which may serve as a foundation for future investigations into perfusion and BBB function in neurodegenerative and other brain disorders.
Collapse
Affiliation(s)
- Xingfeng Shao
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Qinyang Shou
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Kimberly Felix
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Brandon Ojogho
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Xuejuan Jiang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
- Department of Ophthalmology, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Brian T Gold
- Department of Neuroscience, College of Medicine, University of KentuckyFrankfortUnited States
| | - Megan M Herting
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Eric L Goldwaser
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of MedicineBaltimoreUnited States
- Interventional Psychiatry Program, Department of Psychiatry, Weill Cornell MedicineNew YorkUnited States
| | - Peter Kochunov
- Louis A. Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at HoustonHoustonUnited States
| | - Elliot Hong
- Louis A. Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at HoustonHoustonUnited States
| | - Ioannis Pappas
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Meredith Braskie
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Hosung Kim
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Steven Cen
- Department of Radiology and Neurology, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Kay Jann
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Danny JJ Wang
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
- Department of Radiology and Neurology, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| |
Collapse
|
2
|
Almutairi JA, Kidd EJ. Biological Sex Disparities in Alzheimer's Disease. Curr Top Behav Neurosci 2024. [PMID: 39485650 DOI: 10.1007/7854_2024_545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Alzheimer's disease is a highly complex and multifactorial neurodegenerative disorder, with age being the most significant risk factor. The incidence of Alzheimer's disease doubles every 5 years after the age of 65. Consequently, one of the major challenges in Alzheimer's disease research is understanding how the brain changes with age. Gaining insights into these changes could help identify individuals who are more prone to developing Alzheimer's disease as they age. Over the past 25 years, studies on brain aging have examined thousands of human brains to explore the neuronal basis of age-related cognitive decline. However, most of these studies have focused on adults over 60, often neglecting the critical menopause transition period. During menopause, women experience a substantial decline in ovarian sex hormone production, with a decrease of about 90% in estrogen levels. Estrogen is known for its neuroprotective effects, and its significant loss during menopause affects various biological systems, including the brain. Importantly, despite known differences in dementia risk between sexes, the impact of biological sex and sex hormones on brain aging and the development of Alzheimer's disease remains underexplored.
Collapse
Affiliation(s)
- Jawza A Almutairi
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
- Department of Pharmaceutical Science, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Emma J Kidd
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK.
| |
Collapse
|
3
|
Kaczmarczyk M, Deuter CE, Deus H, Kallidou A, Merz CJ, Hellmann-Regen J, Otte C, Wingenfeld K. Effects of separate and combined estradiol and progesterone administration on fear extinction in healthy pre-menopausal women. Transl Psychiatry 2024; 14:449. [PMID: 39448569 PMCID: PMC11502897 DOI: 10.1038/s41398-024-03079-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 10/26/2024] Open
Abstract
Altered fear conditioning and extinction learning are discussed as key etiological features in anxiety disorders. Women have an increased risk for anxiety disorders and fear conditioning has been shown to be influenced by the menstrual cycle phase and circulating gonadal hormones. The objective of our study was to investigate the effects of separate and combined estradiol and progesterone administration on fear extinction in healthy women. We conducted a placebo-controlled, randomized study in healthy women, who completed a fear conditioning paradigm on three consecutive days: fear acquisition training on day 1, fear extinction training on day 2, and return of fear test on day 3. Skin conductance responses (SCRs) served as main outcome variable. Two hours before testing on day 2, participants received pills containing either placebo, estradiol (2 mg), progesterone (400 mg) or the combination of both. We examined 116 women (mean age 25.7 ± 6.0 years), who showed significantly stronger conditioned SCRs to the CS+ than CS- during fear acquisition training indicating successful fear learning. At the beginning of the fear extinction training, estradiol administration reduced the differentiation between the conditioned stimuli. In the return of fear test, the estradiol groups showed heightened SCR responses to the previously extinguished stimulus, i.e., impaired extinction recall. Administration of progesterone did not have any significant influence on SCRs. There were also no effects on fear potentiated startle response. In our interpretation, exogenous estradiol administration affected the extinction of the conditioned fear response which led subsequently to a stronger return of fear. From a clinical perspective our findings suggest that estradiol levels may have an influence on the success of exposure therapy and could be taken into consideration when planning exposure sessions.
Collapse
Affiliation(s)
- Michael Kaczmarczyk
- Department of Psychiatry and Neurosciences, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- BIH Biomedical Innovation Academy, Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Christian Eric Deuter
- Department of Psychiatry and Neurosciences, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Hanna Deus
- Department of Psychiatry and Neurosciences, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anna Kallidou
- Department of Psychiatry and Neurosciences, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christian J Merz
- Department of Cognitive Psychology, Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany
| | - Julian Hellmann-Regen
- Department of Psychiatry and Neurosciences, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christian Otte
- Department of Psychiatry and Neurosciences, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZPG (German Center for Mental Health), partner site Berlin, Berlin, Germany
| | - Katja Wingenfeld
- Department of Psychiatry and Neurosciences, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DZPG (German Center for Mental Health), partner site Berlin, Berlin, Germany
| |
Collapse
|
4
|
Zhang Y, Fletcher JM. Research Note: The Association Between Parity and Odds of Alzheimer's Disease and Dementias Status. Demography 2024; 61:1339-1350. [PMID: 39330980 DOI: 10.1215/00703370-11585876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
While parity is a significant factor influencing parental health, its relationship with dementia remains underexplored. This research note advances the literature by conducting a well-powered analysis of associations between parity (i.e., number of children) and Alzheimer's disease and dementias (AD/D) status in large-scale population data. The data contain a large number of AD/D cases (37,228 women and 19,846 men), allowing a range (1-10) of parity associations to be estimated precisely. Using proxy (adult child's) reports of parental AD/D status, we find that both fathers and mothers with grand multiparity have decreased odds of AD/D status, and the effect sizes become larger as parity increases, with 30-40% reduction in AD/D status at parities above 7. The association is stronger for mothers than for fathers. This finding differs from much of the prior literature and likely suggests the impact of parity, as one of the important life course contexts, on people's cognitive function and risk of having AD/D. Finally, we include population projections that consider how large changes in parity distributions over time may contribute to small elevations in AD/D rates.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Sociology, East Carolina University, Greenville, NC, USA
| | - Jason M Fletcher
- Center for Demography and Ecology, La Follette School of Public Affairs, Department of Population Health Science, and Department of Agricultural and Applied Economics, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
5
|
Kniffin AR, Briand LA. Sex differences in glutamate transmission and plasticity in reward related regions. Front Behav Neurosci 2024; 18:1455478. [PMID: 39359325 PMCID: PMC11445661 DOI: 10.3389/fnbeh.2024.1455478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/30/2024] [Indexed: 10/04/2024] Open
Abstract
Disruptions in glutamate homeostasis within the mesolimbic reward circuitry may play a role in the pathophysiology of various reward related disorders such as major depressive disorders, anxiety, and substance use disorders. Clear sex differences have emerged in the rates and symptom severity of these disorders which may result from differing underlying mechanisms of glutamatergic signaling. Indeed, preclinical models have begun to uncover baseline sex differences throughout the brain in glutamate transmission and synaptic plasticity. Glutamatergic synaptic strength can be assessed by looking at morphological features of glutamatergic neurons including spine size, spine density, and dendritic branching. Likewise, electrophysiology studies evaluate properties of glutamatergic neurons to provide information of their functional capacity. In combination with measures of glutamatergic transmission, synaptic plasticity can be evaluated using protocols that induce long-term potentiation or long-term depression. This review will consider preclinical rodent literature directly comparing glutamatergic transmission and plasticity in reward related regions of males and females. Additionally, we will suggest which regions are exhibiting evidence for sexually dimorphic mechanisms, convergent mechanisms, or no sex differences in glutamatergic transmission and plasticity and highlight gaps in the literature for future investigation.
Collapse
Affiliation(s)
- Alyssa R. Kniffin
- Department of Psychology & Neuroscience, Temple University, Philadelphia, PA, United States
| | - Lisa A. Briand
- Department of Psychology & Neuroscience, Temple University, Philadelphia, PA, United States
- Neuroscience Program, Temple University, Philadelphia, PA, United States
| |
Collapse
|
6
|
Konishi K, Jacobs EG, Aroner S, De Vivo I, Smith B, Scribner-Weiss B, Makris N, Seitz-Holland J, Remington A, Aizley H, Kubicki M, Goldstein JM. Leukocyte telomere length and memory circuitry and cognition in early aging: Impact of sex and menopausal status. Horm Behav 2024; 165:105631. [PMID: 39232410 PMCID: PMC11438173 DOI: 10.1016/j.yhbeh.2024.105631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/06/2024]
Abstract
Telomere length (TL) is an important cellular marker of biological aging impacting the brain and heart. However, how it is related to the brain (e.g., cognitive function and neuroanatomic architecture), and how these relationships may vary by sex and reproductive status, is not well established. Here we assessed the association between leukocyte TL and memory circuitry regional brain volumes and memory performance in early midlife, in relation to sex and reproductive status. Participants (N = 198; 95 females, 103 males; ages 45-55) underwent structural MRI and neuropsychological assessments of verbal, associative, and working memory. Overall, shorter TL was associated with smaller white matter volume in the parahippocampal gyrus and dorsolateral prefrontal cortex. In males, shorter TL was associated with worse working memory performance and corresponding smaller white matter volumes in the parahippocampal gyrus, anterior cingulate cortex, and dorsolateral prefrontal cortex. In females, the impact of cellular aging was revealed over the menopausal transition. In postmenopausal females, shorter TL was associated with poor associative memory performance and smaller grey matter volume in the right hippocampus. In contrast, TL was not related to memory performance or grey and white matter volumes in any memory circuitry region in pre/perimenopausal females. Results demonstrated that shorter TL is associated with worse memory function and smaller volume in memory circuitry regions in early midlife, an association that differs by sex and reproductive status. Taken together, TL may serve as an early indicator of sex-dependent brain abnormalities in early midlife.
Collapse
Affiliation(s)
- Kyoko Konishi
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, United States of America; Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA 02114, United States of America
| | - Emily G Jacobs
- Department of Psychological and Brain Sciences, University of California, Santa Barbara 93111, United States of America
| | - Sarah Aroner
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, United States of America; Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA 02114, United States of America
| | - Immaculata De Vivo
- Department of Epidemiology, T.H. Chan School of Public Health, Boston, MA 02120, United States of America
| | - Brianna Smith
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, United States of America
| | - Blair Scribner-Weiss
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, United States of America
| | - Nikos Makris
- Harvard Medical School, Boston, MA 02120, United States of America; Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, United States of America
| | - Johanna Seitz-Holland
- Harvard Medical School, Boston, MA 02120, United States of America; Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, United States of America
| | - Anne Remington
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, United States of America; Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA 02114, United States of America
| | - Harlyn Aizley
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, United States of America; Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA 02114, United States of America
| | - Marek Kubicki
- Harvard Medical School, Boston, MA 02120, United States of America; Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, United States of America
| | - Jill M Goldstein
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, United States of America; Harvard Medical School, Boston, MA 02120, United States of America; Athinoula A. Martinos Center, Massachusetts General Hospital, Charlestown, MA 02129, United States of America; Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, Boston, MA 02114, United States of America; Department of Medicine, Harvard Medical School, Boston, MA 02115, United States of America.
| |
Collapse
|
7
|
Küchenhoff S, Bayrak Ş, Zsido RG, Saberi A, Bernhardt BC, Weis S, Schaare HL, Sacher J, Eickhoff S, Valk SL. Relating sex-bias in human cortical and hippocampal microstructure to sex hormones. Nat Commun 2024; 15:7279. [PMID: 39179555 PMCID: PMC11344136 DOI: 10.1038/s41467-024-51459-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 07/25/2024] [Indexed: 08/26/2024] Open
Abstract
Determining sex-bias in brain structure is of great societal interest to improve diagnostics and treatment of brain-related disorders. So far, studies on sex-bias in brain structure predominantly focus on macro-scale measures, and often ignore factors determining this bias. Here we study sex-bias in cortical and hippocampal microstructure in relation to sex hormones. Investigating quantitative intracortical profiling in-vivo using the T1w/T2w ratio in 1093 healthy females and males of the cross-sectional Human Connectome Project young adult sample, we find that regional cortical and hippocampal microstructure differs between males and females and that the effect size of this sex-bias varies depending on self-reported hormonal status in females. Microstructural sex-bias and expression of sex hormone genes, based on an independent post-mortem sample, are spatially coupled. Lastly, sex-bias is most pronounced in paralimbic areas, with low laminar complexity, which are predicted to be most plastic based on their cytoarchitectural properties. Albeit correlative, our study underscores the importance of incorporating sex hormone variables into the investigation of brain structure and plasticity.
Collapse
Affiliation(s)
- Svenja Küchenhoff
- Institute of Neuroscience and Medicine (INM-7: Brain and Behavior), Research Centre Jülich, Jülich, Germany.
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
- Institute of Systems Neuroscience, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, UK.
| | - Şeyma Bayrak
- Institute of Neuroscience and Medicine (INM-7: Brain and Behavior), Research Centre Jülich, Jülich, Germany
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Rachel G Zsido
- Cognitive Neuroendocrinology, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Amin Saberi
- Institute of Neuroscience and Medicine (INM-7: Brain and Behavior), Research Centre Jülich, Jülich, Germany
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
- Institute of Systems Neuroscience, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | | | - Susanne Weis
- Institute of Neuroscience and Medicine (INM-7: Brain and Behavior), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - H Lina Schaare
- Institute of Neuroscience and Medicine (INM-7: Brain and Behavior), Research Centre Jülich, Jülich, Germany
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Julia Sacher
- Centre for Integrative Women's Health and Gender Medicine, Medical Faculty & University Hospital Leipzig, Leipzig, Germany
| | - Simon Eickhoff
- Institute of Neuroscience and Medicine (INM-7: Brain and Behavior), Research Centre Jülich, Jülich, Germany
- Institute of Systems Neuroscience, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Sofie L Valk
- Institute of Neuroscience and Medicine (INM-7: Brain and Behavior), Research Centre Jülich, Jülich, Germany.
- Otto Hahn Group Cognitive Neurogenetics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
- Institute of Systems Neuroscience, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
8
|
Khunphet P, Kumarnsit E, Issuriya A, Cheaha D. Estrogen deficiency affects synchronized neural connectivity in the olfactory bulb-nucleus accumbens circuit: A local field potential study in ovariectomized mouse model. Horm Behav 2024; 164:105587. [PMID: 38905819 DOI: 10.1016/j.yhbeh.2024.105587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 05/15/2024] [Accepted: 06/10/2024] [Indexed: 06/23/2024]
Abstract
Estrogen plays a crucial role in regulating various brain functions, including cognitive, emotional, and social behaviors. Menopausal women face a decline in estrogen levels, which has been linked to several physical and mental health issues. However, the impact of estrogen on the olfactory bulb-nucleus accumbens (OB-NAc) circuit, which is essential for regulating emotions and cognitive behaviors, remains poorly understood. To test the hypothesis that estrogen deficiency affects signal processing, we recorded local field potentials (LFPs) using intracranial electrodes implanted in four-week-old ovariectomized (OVX) mice during an open-field test (OFT). The results showed a decrease in locomotor activity and increase in anxiety-like behaviors in OVX mice. Furthermore, we found a decrease in high-gamma power in the OB. We analyzed coherence and inter-region phase-amplitude coupling (ir-PAC) to explore the connectivity between the OB and NAc. We observed a decrease in low-gamma and high-gamma coherence in OVX mice. Additionally, we found that the direction of connectivity from the NAc to the OB was disrupted in OVX mice. In summary, our study provides evidence that estrogen deficiency is linked to synchronized neural connectivity changes in the OB-NAc circuit. These findings have implications for our understanding of the roles played by the OB-NAc neural circuit and estrogen in the regulation of general exploratory behavior and anxiety-like behavior.
Collapse
Affiliation(s)
- Pakavarin Khunphet
- Biology Program, Division of Biological Sciences, Faculty of Science, Prince of Songkla University (PSU), Hatyai, Songkhla, 90110, Thailand; Biosignal Research Center for Health, Faculty of Science, Prince of Songkla University (PSU), Hatyai, Songkhla, 90110, Thailand
| | - Ekkasit Kumarnsit
- Physiology Program, Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University (PSU), Hatyai, Songkhla, 90110, Thailand; Biosignal Research Center for Health, Faculty of Science, Prince of Songkla University (PSU), Hatyai, Songkhla, 90110, Thailand
| | - Acharaporn Issuriya
- Physiology Program, Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University (PSU), Hatyai, Songkhla, 90110, Thailand; Biosignal Research Center for Health, Faculty of Science, Prince of Songkla University (PSU), Hatyai, Songkhla, 90110, Thailand
| | - Dania Cheaha
- Biology Program, Division of Biological Sciences, Faculty of Science, Prince of Songkla University (PSU), Hatyai, Songkhla, 90110, Thailand; Biosignal Research Center for Health, Faculty of Science, Prince of Songkla University (PSU), Hatyai, Songkhla, 90110, Thailand.
| |
Collapse
|
9
|
Jeng SR, Wu GC, Yueh WS, Liu PH, Kuo SF, Dufour S, Chang CF. The expression profiles of cyp19a1, sf-1, esrs and gths in the brain-pituitary during gonadal sex differentiation in juvenile Japanese eels. Gen Comp Endocrinol 2024; 353:114512. [PMID: 38582176 DOI: 10.1016/j.ygcen.2024.114512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/08/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
Eels are gonochoristic species whose gonadal differentiation initiates at the yellow eel stage and is influenced by environmental factors. We revealed some sex-related genes were sex dimorphically expressed in gonads during gonadal sex differentiation of Japanese eel (Anguilla japonica); however, the expression of sex-related genes in the brain-pituitary during gonadal sex differentiation in eels is still unclear. This study aimed to investigate the sex-related gene expressions in the brain-pituitary and tried to clarify their roles in the brain and gonads during gonadal sex differentiation. Based on our previous histological study, the control eels developed as males, and estradiol-17β (E2) was used for feminization. Our results showed that during testicular differentiation, the brain cyp19a1 transcripts and aromatase proteins were increased significantly; moreover, the cyp19a1, sf-1, foxl2s, and esrs (except gperb) transcripts in the midbrain/pituitary also were increased significantly. Forebrain gnrh1 transcripts increased slightly during gonadal differentiation of both sexes, but the gnrhr1b and gnrhr2 transcripts in the midbrain/pituitary were stable during gonadal differentiation. The expression levels of gths and gh in the midbrain/pituitary were significantly increased during testicular differentiation and were much higher in males than in E2-feminized females. These results implied that endogenous estrogens might play essential roles in the brain/pituitary during testicular differentiation, sf-1, foxl2s, and esrs may have roles in cyp19a1 regulation in the midbrain/pituitary of Japanese eels. For the GnRH-GTH axis, gths, especially fshb, may be regulated by esrs and involved in regulating testicular differentiation and development in Japanese eels.
Collapse
Affiliation(s)
- Shan-Ru Jeng
- Department of Aquaculture, National Kaohsiung University of Science and Technology, Kaohsiung 811, Taiwan.
| | - Guan-Chung Wu
- Department of Aquaculture, National Taiwan Ocean University, Keelung 202, Taiwan.
| | - Wen-Shiun Yueh
- Department of Aquaculture, National Kaohsiung University of Science and Technology, Kaohsiung 811, Taiwan
| | - Pei-Hua Liu
- Department of Aquaculture, National Kaohsiung University of Science and Technology, Kaohsiung 811, Taiwan
| | - Shu-Fen Kuo
- Department of Aquaculture, National Kaohsiung University of Science and Technology, Kaohsiung 811, Taiwan
| | - Sylvie Dufour
- Biology of Aquatic Organisms and Ecosystems (BOREA), Muséum National d'Histoire Naturelle, Sorbonne Université, CNRS, IRD, Paris, France; Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung 202, Taiwan
| | - Ching-Fong Chang
- Department of Aquaculture, National Taiwan Ocean University, Keelung 202, Taiwan; Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung 202, Taiwan.
| |
Collapse
|
10
|
Yuan M, Rong M, Long X, Lian S, Fang Y. Trajectories of cognitive decline in different domains prior to AD onset in persons with mild cognitive impairment. Arch Gerontol Geriatr 2024; 122:105375. [PMID: 38431989 DOI: 10.1016/j.archger.2024.105375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/29/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVES To explore the trajectories and the change-points of global and five domain-specific cognitive functions before the onset of Alzheimer's disease (AD). METHODS Data was retrieved from the Alzheimer's Disease Neuroimaging Initiative with follow-up from 2005 to 2022. Participants with mild cognitive impairment (MCI) at baseline and those who progressed to AD during follow-up were included. The time of AD onset was defined as the visit time when participant was first diagnosed as AD during follow-up. Global and five domain-specific cognitive functions (immediate memory, visuospatial ability, language, processing speed and executive function) were assessed by Mini-Mental State Examination, Immediate recalling trials of Rey Auditory Verbal Learning Test, Clock Drawing Test, Animal Fluency Test, Part A and B of Trail Making Test, respectively. Their trajectories and change-points before AD onset were explored by generalized additive mixed models and piecewise linear regression models, respectively. RESULTS 349 participants were diagnosed as MCI at baseline and converted to AD during follow-up, who were included in this study. They had been visited on an average of 4.6 times (SD = 2.1, range = 2.0-13.0), with a total of 1593 visits. Their mean baseline age and AD onset age were 74.4 (SD = 6.4, range = 60.0-88.4) and 77.0 (SD = 6.8, range = 60.5-94.7) years, respectively. Baseline age and educational year were significantly associated with global cognitive, immediate memory, language and executive function. Men presented better global cognitive function (β = 0.54, p < 0.05) but poorer immediate memory (β = -1.72, p < 0.05) than women. Immediate memory and visuospatial ability showed the earliest change-points at 4 years before the onset of AD (Note as T-4years), followed by language (T-3.5years), executive function (T-2.5 years), processing speed (T-2.0 years), and finally the global cognitive function (T-1.5years). CONCLUSIONS The trajectories of the six neuropsychological scores were non-linear and showed deterioration in functions over time. Immediate memory and visuospatial ability showed the earliest change-points prior to AD onset.
Collapse
Affiliation(s)
- Manqiong Yuan
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China; Key Laboratory of Health Technology Assessment of Fujian Province, School of Public Health, Xiamen University, China
| | - Meng Rong
- Key Laboratory of Health Technology Assessment of Fujian Province, School of Public Health, Xiamen University, China
| | - Xianxian Long
- Key Laboratory of Health Technology Assessment of Fujian Province, School of Public Health, Xiamen University, China
| | - Shuli Lian
- Key Laboratory of Health Technology Assessment of Fujian Province, School of Public Health, Xiamen University, China
| | - Ya Fang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, China; Key Laboratory of Health Technology Assessment of Fujian Province, School of Public Health, Xiamen University, China.
| |
Collapse
|
11
|
Shao X, Shou Q, Felix K, Ojogho B, Jiang X, Gold BT, Herting MM, Goldwaser EL, Kochunov P, Hong LE, Pappas I, Braskie M, Kim H, Cen S, Jann K, Wang DJJ. Age-Related Decline in Blood-Brain Barrier Function is More Pronounced in Males than Females in Parietal and Temporal Regions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.12.575463. [PMID: 38293052 PMCID: PMC10827081 DOI: 10.1101/2024.01.12.575463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The blood-brain barrier (BBB) plays a pivotal role in protecting the central nervous system (CNS), shielding it from potential harmful entities. A natural decline of BBB function with aging has been reported in both animal and human studies, which may contribute to cognitive decline and neurodegenerative disorders. Limited data also suggest that being female may be associated with protective effects on BBB function. Here we investigated age and sex-dependent trajectories of perfusion and BBB water exchange rate (kw) across the lifespan in 186 cognitively normal participants spanning the ages of 8 to 92 years old, using a non-invasive diffusion prepared pseudo-continuous arterial spin labeling (DP-pCASL) MRI technique. We found that the pattern of BBB kw decline with aging varies across brain regions. Moreover, results from our DP-pCASL technique revealed a remarkable decline in BBB kw beginning in the early 60s, which was more pronounced in males. In addition, we observed sex differences in parietal and temporal regions. Our findings provide in vivo results demonstrating sex differences in the decline of BBB function with aging, which may serve as a foundation for future investigations into perfusion and BBB function in neurodegenerative and other brain disorders.
Collapse
Affiliation(s)
- Xingfeng Shao
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Qinyang Shou
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Kimberly Felix
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California
| | - Brandon Ojogho
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Xuejuan Jiang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California
- Department of Ophthalmology, Keck School of Medicine, University of Southern California
| | - Brian T. Gold
- Department of Neuroscience, College of Medicine, University of Kentucky
| | - Megan M Herting
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California
| | - Eric L Goldwaser
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine
- Interventional Psychiatry Program, Department of Psychiatry, Weill Cornell Medicine
| | - Peter Kochunov
- Louis A. Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston
| | - L. Elliot Hong
- Louis A. Faillace Department of Psychiatry and Behavioral Sciences at McGovern Medical School, The University of Texas Health Science Center at Houston
| | - Ioannis Pappas
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Meredith Braskie
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Hosung Kim
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Steven Cen
- Department of Radiology and Neurology, Keck School of Medicine, University of Southern California
| | - Kay Jann
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
| | - Danny JJ Wang
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California
- Department of Radiology and Neurology, Keck School of Medicine, University of Southern California
| |
Collapse
|
12
|
Gottschewsky N, Kraft D, Kaufmann T. Menarche, pubertal timing and the brain: female-specific patterns of brain maturation beyond age-related development. Biol Sex Differ 2024; 15:25. [PMID: 38532493 PMCID: PMC10964568 DOI: 10.1186/s13293-024-00604-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 03/18/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Puberty depicts a period of profound and multifactorial changes ranging from social to biological factors. While brain development in youths has been studied mostly from an age perspective, recent evidence suggests that pubertal measures may be more sensitive to study adolescent neurodevelopment, however, studies on pubertal timing in relation to brain development are still scarce. METHODS We investigated if pre- vs. post-menarche status can be classified using machine learning on cortical and subcortical structural magnetic resonance imaging (MRI) data from strictly age-matched adolescent females from the Adolescent Brain Cognitive Development (ABCD) cohort. For comparison of the identified menarche-related patterns to age-related patterns of neurodevelopment, we trained a brain age prediction model on data from the Philadelphia Neurodevelopmental Cohort and applied it to the same ABCD data, yielding differences between predicted and chronological age referred to as brain age gaps. We tested the sensitivity of both these frameworks to measures of pubertal maturation, specifically age at menarche and puberty status. RESULTS The machine learning model achieved moderate but statistically significant accuracy in the menarche classification task, yielding for each subject a class probability ranging from 0 (pre-) to 1 (post- menarche). Comparison to brain age predictions revealed shared and distinct patterns of neurodevelopment captured by both approaches. Continuous menarche class probabilities were positively associated with brain age gaps, but only the menarche class probabilities-not the brain age gaps-were associated with age at menarche. CONCLUSIONS This study demonstrates the use of a machine learning model to classify menarche status from structural MRI data while accounting for age-related neurodevelopment. Given its sensitivity towards measures of puberty timing, our work suggests that menarche class probabilities may be developed toward an objective brain-based marker of pubertal development.
Collapse
Affiliation(s)
- Nina Gottschewsky
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, Tübingen, Germany.
- Max Planck School of Cognition, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany.
| | - Dominik Kraft
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, Tübingen, Germany
| | - Tobias Kaufmann
- Department of Psychiatry and Psychotherapy, Tübingen Center for Mental Health, University of Tübingen, Tübingen, Germany.
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- German Center for Mental Health (DZPG), Partner Site Tübingen, Tübingen, Germany.
| |
Collapse
|
13
|
Spets DS, Cohen JE, Konishi K, Aroner S, Misra M, Lee H, Goldstein JM. Impact of sex and reproductive status on the default mode network in early midlife: implications for aging of memory circuitry and function. Cereb Cortex 2024; 34:bhae088. [PMID: 38494419 PMCID: PMC10944696 DOI: 10.1093/cercor/bhae088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 03/19/2024] Open
Abstract
Alterations to the resting-state default mode network (rsDMN) are early indicators of memory decline and Alzheimer's disease (AD). Brain regions shared by the rsDMN and memory circuitry are highly sexually dimorphic. However, data are limited regarding the impact of sex and reproductive status on rsDMN connectivity and memory circuitry and function. In the current investigation, rsDMN connectivity was assessed in 180 early midlife adults aged 45 to 55 by sex and reproductive status (87 women; 93 men). Associations between left and right hippocampal connectivity of rsDMN and verbal memory encoding circuitry were examined using linear mixed models, controlled for age and parental socioeconomic status, testing interactions by sex and reproductive status. Relative to men, women exhibited greater rsDMN connectivity between the left and right hippocampus. In relation to rsDMN-memory encoding connectivity, sex differences were revealed across the menopausal transition, such that only postmenopausal women exhibited loss of the ability to decrease rsDMN left-right hippocampal connectivity during memory encoding associated with poorer memory performance. Results demonstrate that sex and reproductive status play an important role in aging of the rsDMN and interactions with memory circuitry/function. This suggests the critical importance of sex and reproductive status when studying early midlife indicators of memory decline and AD risk.
Collapse
Affiliation(s)
- Dylan S Spets
- Clinical Neuroscience Laboratory for Sex Differences in the Brain, Department of Psychiatry, Massachusetts General Hospital, 149 13th Street, Boston, MA 02129, USA
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
- Department of Psychiatry, Harvard Medical School, 401 Park Drive, Boston, MA 02215, USA
| | - Justine E Cohen
- Clinical Neuroscience Laboratory for Sex Differences in the Brain, Department of Psychiatry, Massachusetts General Hospital, 149 13th Street, Boston, MA 02129, USA
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
| | - Kyoko Konishi
- Clinical Neuroscience Laboratory for Sex Differences in the Brain, Department of Psychiatry, Massachusetts General Hospital, 149 13th Street, Boston, MA 02129, USA
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
- Department of Psychiatry, Harvard Medical School, 401 Park Drive, Boston, MA 02215, USA
| | - Sarah Aroner
- Clinical Neuroscience Laboratory for Sex Differences in the Brain, Department of Psychiatry, Massachusetts General Hospital, 149 13th Street, Boston, MA 02129, USA
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
| | - Madhusmita Misra
- Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
- Department of Pediatrics, Division of Pediatric Endocrinology, Massachusetts General Hospital, 55 Fruit Street Boston, MA 02114, USA
| | - Hang Lee
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
- Biostatistics Center, Massachusetts General Hospital, 500 Staniford Street, Boston, MA 02114, USA
| | - Jill M Goldstein
- Clinical Neuroscience Laboratory for Sex Differences in the Brain, Department of Psychiatry, Massachusetts General Hospital, 149 13th Street, Boston, MA 02129, USA
- Innovation Center on Sex Differences in Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
- Department of Psychiatry, Harvard Medical School, 401 Park Drive, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| |
Collapse
|
14
|
Schweitzer N, Li J, Thurston RC, Lopresti B, Klunk WE, Snitz B, Tudorascu D, Cohen A, Kamboh MI, Halligan‐Eddy E, Iordanova B, Villemagne VL, Aizenstein H, Wu M. Sex-dependent alterations in hippocampal connectivity are linked to cerebrovascular and amyloid pathologies in normal aging. Alzheimers Dement 2024; 20:914-924. [PMID: 37817668 PMCID: PMC10916980 DOI: 10.1002/alz.13503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/29/2023] [Accepted: 09/19/2023] [Indexed: 10/12/2023]
Abstract
INTRODUCTION Compared to males, females have an accelerated trajectory of cognitive decline in Alzheimer's disease (AD). The neurobiological factors underlying the more rapid cognitive decline in AD in females remain unclear. This study explored how sex-dependent alterations in hippocampal connectivity over 2 years are associated with cerebrovascular and amyloid pathologies in normal aging. METHODS Thirty-three females and 21 males 65 to 93 years of age with no cognitive impairment performed a face-name associative memory functional magnetic resonance imaging (fMRI) task with a 2-year follow-up. We acquired baseline carbon 11-labeled Pittsburgh compound B ([11 C]PiB) positron emission tomography (PET) and T2-weighted fluid-attenuated inversion recovery (T2-FLAIR) MRI to quantify amyloid β (Aβ) burden and white matter hyperintensity (WMH) volume, respectively. RESULTS Males had increased hippocampal-prefrontal connectivity over 2 years, associated with greater Aβ burden. Females had increased bilateral hippocampal functional connectivity, associated with greater WMH volume. DISCUSSION These findings suggest sex-dependent compensatory mechanisms in the memory network in the presence of cerebrovascular and AD pathologies and may explain the accelerated trajectory of cognitive decline in females.
Collapse
Affiliation(s)
- Noah Schweitzer
- Department of BioengineeringUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Jinghang Li
- Department of BioengineeringUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Rebecca C. Thurston
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Brian Lopresti
- Department of RadiologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - William E. Klunk
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Beth Snitz
- Department of NeurologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Dana Tudorascu
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Ann Cohen
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - M. Ilyas Kamboh
- Department of Human GeneticsSchool of Public HealthUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Edythe Halligan‐Eddy
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Bistra Iordanova
- Department of BioengineeringUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Victor L. Villemagne
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Howard Aizenstein
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Minjie Wu
- Department of PsychiatryUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|
15
|
Arifin H, Chen R, Banda KJ, Kustanti CY, Chang CY, Lin HC, Liu D, Lee TY, Chou KR. Meta-analysis and moderator analysis of the prevalence of malnutrition and malnutrition risk among older adults with dementia. Int J Nurs Stud 2024; 150:104648. [PMID: 38043486 DOI: 10.1016/j.ijnurstu.2023.104648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 11/06/2023] [Accepted: 11/16/2023] [Indexed: 12/05/2023]
Abstract
BACKGROUND Aging and dementia are common and closely related health problems in older adults, affecting their ability to maintain a healthy diet and ultimately resulting in malnutrition. OBJECTIVE In this study, we estimated the global prevalence of malnutrition and malnutrition risk in older adults with dementia. DESIGN Meta-analysis. DATA SOURCES Embase, Ovid MEDLINE, PubMed, CINAHL, Scopus, and Web of Science were comprehensively searched for articles published from database inception to October 2022. METHODS Pooled prevalence analysis was conducted using a generalized linear mixed model and a random-effects model. I2 and Cochran's Q statistics were used for identifying heterogeneity. Publication bias was evaluated using Peters' regression test and a funnel plot. Moderator analyses were conducted to investigate variations in the prevalence estimates of the included studies. All statistical analyses were conducted using R software. RESULTS A total of 16 studies involving a total of 6513 older adults with dementia were included in the analysis. The results indicated that 32.52 % (95 % confidence interval: 19.55-45.49) of all included older adults with dementia had malnutrition, whereas 46.80 % (95 % confidence interval: 38.90-54.70) had a risk of malnutrition. The prevalence of malnutrition was found to be high among older patients living in institutionalized settings (46.59 %) and those with Alzheimer's disease (12.26 %). The factors moderating the prevalence of malnutrition included adequate vitamin B12 consumption, risk behaviors, medical comorbidities, and certain neuropsychiatric symptoms. The prevalence of malnutrition risk was high among women (29.84 %) and patients with Alzheimer's disease (26.29 %). The factors moderating the prevalence of malnutrition risk included total cholesterol level, vitamin B12 consumption, risk behaviors, medical comorbidities, and certain neuropsychiatric symptoms. CONCLUSIONS Approximately one-third of older adults with dementia are malnourished and nearly half of older adults are at a risk of malnutrition. Encouraging collaboration among health-care professionals and ensuring early assessment and effective management of malnutrition are crucial for maintaining a favorable nutritional status in older adults with dementia. REGISTRATION This study was registered with the International Prospective Register of Systematic Reviews (PROSPERO: CRD42022369329). TWEETABLE ABSTRACT Globally, approximately 32.52 % of older adults with dementia are malnourished and approximately 46.80 % are at a risk of malnutrition.
Collapse
Affiliation(s)
- Hidayat Arifin
- School of Nursing, College of Nursing, Taipei Medical University, Taipei, Taiwan; Faculty of Nursing, Universitas Airlangga, Surabaya, Indonesia. https://twitter.com/ha_arifin
| | - Ruey Chen
- School of Nursing, College of Nursing, Taipei Medical University, Taipei, Taiwan; Department of Nursing, Taipei Medical University-Shuang Ho Hospital, New Taipei, Taiwan; Post-Baccalaureate Program in Nursing, College of Nursing, Taipei Medical University, Taipei, Taiwan
| | - Kondwani Joseph Banda
- School of Nursing, College of Nursing, Taipei Medical University, Taipei, Taiwan; Endoscopy Unit, Surgery Department, Kamuzu Central Hospital, Lilongwe, Malawi
| | - Christina Yeni Kustanti
- School of Nursing, College of Nursing, Taipei Medical University, Taipei, Taiwan; Sekolah Tinggi Ilmu Kesehatan Bethesda Yakkum, Yogyakarta, Indonesia
| | - Ching-Yi Chang
- School of Nursing, College of Nursing, Taipei Medical University, Taipei, Taiwan
| | - Hui-Chen Lin
- School of Nursing, College of Nursing, Taipei Medical University, Taipei, Taiwan; Research Center in Nursing Clinical Practice, Wan Fang Hospital Taipei Medical University, Taipei, Taiwan
| | - Doresses Liu
- School of Nursing, College of Nursing, Taipei Medical University, Taipei, Taiwan; Research Center in Nursing Clinical Practice, Wan Fang Hospital Taipei Medical University, Taipei, Taiwan; Department of Nursing, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Tso-Ying Lee
- School of Nursing, College of Nursing, Taipei Medical University, Taipei, Taiwan; Nursing Research Center, Department of Nursing, Taipei Medical University Hospital, Taipei, Taiwan
| | - Kuei-Ru Chou
- School of Nursing, College of Nursing, Taipei Medical University, Taipei, Taiwan; Department of Nursing, Taipei Medical University-Shuang Ho Hospital, New Taipei, Taiwan; Research Center in Nursing Clinical Practice, Wan Fang Hospital Taipei Medical University, Taipei, Taiwan; Psychiatric Research Center, Taipei Medical University Hospital, Taipei, Taiwan; Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
16
|
Lipatova O, Campolattaro MM, Lockhart BK, Hammad MB. Differential effects of acute stress on spatial learning and memory in the open-field tower maze across the female estrous cycle. Neurobiol Learn Mem 2023; 206:107862. [PMID: 37944635 DOI: 10.1016/j.nlm.2023.107862] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 10/25/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023]
Abstract
The purpose of the present investigation was to test how acute stress and levels of circulating estrogens together influence acquisition and retention of spatial learning, as well as explorative behaviors in female rats. We used the hippocampus-dependent Open-field Tower Maze (OFTM) task to assess acquisition followed by a retention test (reacquisition) that was given 48 h later. Immediately prior to acquisition, experimental rats were exposed to an acute restraint stress and were trained under bright lights. Female rats' estrous cycles were tracked throughout training and testing. Exposure to stress did not affect learning when levels of estrogens were low (i.e., during estrus and metestrus). However, acute stress exposure significantly lowered spatial acquisition of the female rats in the phases with rising levels of estrogens (i.e., during diestrus and proestrus). Furthermore, this stress-induced diminishment during acquisition was evident at the beginning of the retention without any presentation of stress. The present findings provide insight about the interactive relationship between stress and sex hormones on cognitive functions.
Collapse
Affiliation(s)
- Olga Lipatova
- Christopher Newport University, Newport News, VA, United States.
| | | | - Blakely K Lockhart
- Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Mariam B Hammad
- Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
17
|
Branigan GL, Torrandell-Haro G, Chen S, Shang Y, Perez-Miller S, Mao Z, Padilla-Rodriguez M, Cortes-Flores H, Vitali F, Brinton RD. Breast cancer therapies reduce risk of Alzheimer's disease and promote estrogenic pathways and action in brain. iScience 2023; 26:108316. [PMID: 38026173 PMCID: PMC10663748 DOI: 10.1016/j.isci.2023.108316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/08/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Worldwide, an ever-increasing number of women are prescribed estrogen-modulating therapies (EMTs) for the treatment of breast cancer. In parallel, aging of the global population of women will contribute to risk of both breast cancer and Alzheimer's disease. To address the impact of anti-estrogen therapies on risk of Alzheimer's and neural function, we conducted medical informatic and molecular pharmacology analyses to determine the impact of EMTs on risk of Alzheimer's followed by determination of EMT estrogenic mechanisms of action in neurons. Collectively, these data provide both clinical and mechanistic data indicating that select EMTs exert estrogenic agonist action in neural tissue that are associated with reduced risk of Alzheimer's disease while simultaneously acting as effective estrogen receptor antagonists in breast.
Collapse
Affiliation(s)
- Gregory L. Branigan
- Center for Innovation in Brain Science, University of Arizona; Tucson AZ, USA
- Department of Pharmacology, University of Arizona College of Medicine; Tucson AZ, USA
- Medical Scientist Training Program, University of Arizona College of Medicine; Tucson AZ, USA
| | - Georgina Torrandell-Haro
- Center for Innovation in Brain Science, University of Arizona; Tucson AZ, USA
- Department of Pharmacology, University of Arizona College of Medicine; Tucson AZ, USA
| | - Shuhua Chen
- Center for Innovation in Brain Science, University of Arizona; Tucson AZ, USA
| | - Yuan Shang
- Center for Innovation in Brain Science, University of Arizona; Tucson AZ, USA
| | | | - Zisu Mao
- Center for Innovation in Brain Science, University of Arizona; Tucson AZ, USA
| | | | | | - Francesca Vitali
- Center for Innovation in Brain Science, University of Arizona; Tucson AZ, USA
- Center of Bioinformatics and Biostatistics, University of Arizona College of Medicine; Tucson AZ, USA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, University of Arizona; Tucson AZ, USA
- Department of Pharmacology, University of Arizona College of Medicine; Tucson AZ, USA
- Department of Neurology, University of Arizona College of Medicine; Tucson AZ, USA
| |
Collapse
|
18
|
Aquilani R, Cotta Ramusino M, Maestri R, Iadarola P, Boselli M, Perini G, Boschi F, Dossena M, Bellini A, Buonocore D, Doria E, Costa A, Verri M. Several dementia subtypes and mild cognitive impairment share brain reduction of neurotransmitter precursor amino acids, impaired energy metabolism, and lipid hyperoxidation. Front Aging Neurosci 2023; 15:1237469. [PMID: 37655338 PMCID: PMC10466813 DOI: 10.3389/fnagi.2023.1237469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023] Open
Abstract
Objective Dementias and mild cognitive impairment (MCI) are associated with variously combined changes in the neurotransmitter system and signaling, from neurotransmitter synthesis to synaptic binding. The study tested the hypothesis that different dementia subtypes and MCI may share similar reductions of brain availability in amino acid precursors (AAPs) of neurotransmitter synthesis and concomitant similar impairment in energy production and increase of oxidative stress, i.e., two important metabolic alterations that impact neurotransmission. Materials and methods Sixty-five demented patients (Alzheimer's disease, AD, n = 44; frontotemporal disease, FTD, n = 13; vascular disease, VaD, n = 8), 10 subjects with MCI and 15 control subjects (CTRL) were recruited for this study. Cerebrospinal fluid (CSF) and plasma levels of AAPs, energy substrates (lactate, pyruvate), and an oxidative stress marker (malondialdehyde, MDA) were measured in all participants. Results Demented patients and subjects with MCI were similar for age, anthropometric parameters, biohumoral variables, insulin resistance (HOMA index model), and CSF neuropathology markers. Compared to age-matched CTRL, both demented patients and MCI subjects showed low CSF AAP tyrosine (precursor of dopamine and catecholamines), tryptophan (precursor of serotonin), methionine (precursor of acetylcholine) limited to AD and FTD, and phenylalanine (an essential amino acid largely used for protein synthesis) (p = 0.03 to <0.0001). No significant differences were found among dementia subtypes or between each dementia subtype and MCI subjects. In addition, demented patients and MCI subjects, compared to CTRL, had similar increases in CSF and plasma levels of pyruvate (CSF: p = 0.023 to <0.0001; plasma: p < 0.002 to <0.0001) and MDA (CSF: p < 0.035 to 0.002; plasma: p < 0.0001). Only in AD patients was the CSF level of lactate higher than in CTRL (p = 0.003). Lactate/pyruvate ratios were lower in all experimental groups than in CTRL. Conclusion AD, FTD, and VaD dementia patients and MCI subjects may share similar deficits in AAPs, partly in energy substrates, and similar increases in oxidative stress. These metabolic alterations may be due to AAP overconsumption following high brain protein turnover (leading to phenylalanine reductions), altered mitochondrial structure and function, and an excess of free radical production. All these metabolic alterations may have a negative impact on synaptic plasticity and activity.
Collapse
Affiliation(s)
- Roberto Aquilani
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Matteo Cotta Ramusino
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementia, IRCCS C. Mondino Foundation, Pavia, Italy
- Dementia Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Roberto Maestri
- Department of Biomedical Engineering of the Montescano Institute, Istituti Clinici Scientifici Maugeri IRCCS, Montescano, Italy
| | - Paolo Iadarola
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Mirella Boselli
- Neurorehabilitation Unit of the Montescano Institute, Istituti Clinici Scientifici Maugeri IRCCS, Montescano, Italy
| | - Giulia Perini
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementia, IRCCS C. Mondino Foundation, Pavia, Italy
- Dementia Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Federica Boschi
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Maurizia Dossena
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Anna Bellini
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Daniela Buonocore
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Enrico Doria
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Alfredo Costa
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementia, IRCCS C. Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Manuela Verri
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| |
Collapse
|
19
|
Ponnusamy V, Solaiyappan K, Govindarasu M, Prathap L, Babu S, Krishnan M. Effect of estradiol on histopathology of brain in unilateral and bilateral ovariectomized rats. Bioinformation 2023; 19:703-706. [PMID: 37885784 PMCID: PMC10598359 DOI: 10.6026/97320630019703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/30/2023] [Accepted: 06/30/2023] [Indexed: 10/28/2023] Open
Abstract
Estradiol, a major form of estrogen, plays a crucial role in various aspects of brain function, including neuronal health, synaptic plasticity, and cognitive processes. Therefore, it is of interest to investigate the histopathological changes of the rat brain following estradiol treatment in unilateral and bilateral ovariectomized rats, a commonly used method to induce estrogen deficiency and study the consequences of hormonal changes. Two months old Wistar albino female rats were divided into five experimental groups, each consisting of 6 animals. Unilateral and bilateral ovariectomized rats were treated with estradiol (10 mg/Kg/b.wt) subcutaneously for 30 days. The histopathological analysis of brain revealed normal 5-6 compact layers of small pyramidal cells of CA3 region, most with vesicular nuclei in control rats andan irregular and disturbed thickness of the pyramidal cell layer in the CA3 region, suggesting neuronal loss in unilateral ovariectomized rats. Estradiol therapy to these rats showed dark hyperchromatic layers of pyramidal cells in the CA3 region, most of which displayed vesicular nuclei and less shrinkage. Bilateral ovariectomized rats without estradiol treatment showed irregular and disturbed thickness of the pyramidal cell layer in the CA3 region Similar to Group II, indicating neuronal loss and bilateral ovariectomized rats with estradiol treatment showed no significant changes. These findings highlight the potential role of estradiol in modulating the histopathological changes associated with ovariectomy. Further research is warranted to elucidate the underlying mechanisms through which estradiol exerts its effects on neuronal integrity and to explore potential therapeutic strategies for preventing or reversing these histological changes.
Collapse
Affiliation(s)
- Vaijayanthimala Ponnusamy
- />Research scholar, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamilnadu, India
- />Department of Anatomy,Sri Lalithambigai Medical College and hospital, Chennai, Tamilnadu, India
| | - Kamalakannan Solaiyappan
- />Department of Anatomy, Shri Sathya Sai Medical College and Research Institute, Sri Balaji Vidyapeeth (SBV) - Deemed to be University, Ammapettai, Nellikuppam, Chengalpet District, Tamil Nadu - 603108
| | - Mydhili Govindarasu
- />Biomedical research unit and laboratory animal centre, Department of Anatomy, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamilnadu, India
| | - Lavanya Prathap
- />Department of Anatomy, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamilnadu, India
| | - Shyamaladevi Babu
- />Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam-603103, Tamil Nadu, India
| | - Madhan Krishnan
- />Research, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam-603103, Tamil Nadu, India
| |
Collapse
|
20
|
Ficek-Tani B, Horien C, Ju S, Xu W, Li N, Lacadie C, Shen X, Scheinost D, Constable T, Fredericks C. Sex differences in default mode network connectivity in healthy aging adults. Cereb Cortex 2023; 33:6139-6151. [PMID: 36563018 PMCID: PMC10183749 DOI: 10.1093/cercor/bhac491] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/18/2022] [Accepted: 11/20/2022] [Indexed: 12/24/2022] Open
Abstract
Women show an increased lifetime risk of Alzheimer's disease (AD) compared with men. Characteristic brain connectivity changes, particularly within the default mode network (DMN), have been associated with both symptomatic and preclinical AD, but the impact of sex on DMN function throughout aging is poorly understood. We investigated sex differences in DMN connectivity over the lifespan in 595 cognitively healthy participants from the Human Connectome Project-Aging cohort. We used the intrinsic connectivity distribution (a robust voxel-based metric of functional connectivity) and a seed connectivity approach to determine sex differences within the DMN and between the DMN and whole brain. Compared with men, women demonstrated higher connectivity with age in posterior DMN nodes and lower connectivity in the medial prefrontal cortex. Differences were most prominent in the decades surrounding menopause. Seed-based analysis revealed higher connectivity in women from the posterior cingulate to angular gyrus, which correlated with neuropsychological measures of declarative memory, and hippocampus. Taken together, we show significant sex differences in DMN subnetworks over the lifespan, including patterns in aging women that resemble changes previously seen in preclinical AD. These findings highlight the importance of considering sex in neuroimaging studies of aging and neurodegeneration.
Collapse
Affiliation(s)
- Bronte Ficek-Tani
- Department of Neurology, Yale School of Medicine, New Haven, CT 06520, United States
| | - Corey Horien
- Interdepartmental Neuroscience Program, Yale School of Medicine, New Haven, CT 06520, United States
| | - Suyeon Ju
- Department of Neurology, Yale School of Medicine, New Haven, CT 06520, United States
| | - Wanwan Xu
- Department of Biostatistics, Yale School of Medicine, New Haven, CT 06520, United States
| | - Nancy Li
- Department of Neurology, Yale School of Medicine, New Haven, CT 06520, United States
| | - Cheryl Lacadie
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, United States
| | - Xilin Shen
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, United States
| | - Dustin Scheinost
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, United States
| | - Todd Constable
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT 06520, United States
| | - Carolyn Fredericks
- Department of Neurology, Yale School of Medicine, New Haven, CT 06520, United States
| |
Collapse
|
21
|
Zhang Y, Fletcher J, Lu Q, Song J. Gender differences in the association between parity and cognitive function: Evidence from the UK biobank. Soc Sci Med 2023; 320:115649. [PMID: 36709690 PMCID: PMC9974636 DOI: 10.1016/j.socscimed.2022.115649] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/29/2022] [Accepted: 12/31/2022] [Indexed: 01/05/2023]
Abstract
While much previous work linking fertility history with late-life cognition has focused on a narrow set of cognitive measures and/or has used modest sample sizes in the analysis, our paper expands the size and scope of these linkages by analyzing cognitive function across five domains and precisely estimating gendered patterns between men and women. Results point to important gendered associations between parity and cognition: having children is likely associated with better cognitive function for fathers in all five domains. However, mothers show worse cognitive function in some domains (i.e., numeric memory, prospective memory, and fluid intelligence) than childless women. We explore the possibility of confounding in these associations and rule out the effects of genetic cognitive ability on fertility. We also find that adding controls for educational attainment differ by gender-strengthening associations between parity and cognition for men and largely eliminating them for women. The findings support previous work done on how life course contexts may link to the risk of dementia or cognitive impairment, highlighting parity as potential protective or risk factors to parents' cognitive health. The use of five cognitive domains yields variations in results, giving implications on measure selection of cognitive function and calling for replicated work covering more cognitive domains.
Collapse
Affiliation(s)
- Yan Zhang
- Center for Demography of Health and Aging, University of Wisconsin, Madison, United States.
| | - Jason Fletcher
- Center for Demography of Health and Aging, University of Wisconsin, Madison, United States.
| | - Qiongshi Lu
- Department of Biostatistics and Medical Informatics, University of Wisconsin, Madison, United States.
| | - Jie Song
- Department of Statistics, University of Wisconsin, Madison, United States.
| |
Collapse
|
22
|
Williams VJ, Koscik R, Sicinski K, Johnson SC, Herd P, Asthana S. Associations Between Midlife Menopausal Hormone Therapy Use, Incident Diabetes, and Late Life Memory in the Wisconsin Longitudinal Study. J Alzheimers Dis 2023; 93:727-741. [PMID: 37092221 PMCID: PMC10551825 DOI: 10.3233/jad-221240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
BACKGROUND Prior research suggests a link between menopausal hormone therapy (MHT) use, memory function, and diabetes risk. The menopausal transition is a modifiable period to enhance long-term health and cognitive outcomes, although studies have been limited by short follow-up periods precluding a solid understanding of the lasting effects of MHT use on cognition. OBJECTIVE We examined the effects of midlife MHT use on subsequent diabetes incidence and late life memory performance in a large, same-aged, population-based cohort. We hypothesized that the beneficial effects of MHT use on late life cognition would be partially mediated by reduced diabetes risk. METHODS 1,792 women from the Wisconsin Longitudinal Study (WLS) were included in analysis. We employed hierarchical linear regression, Cox regression, and causal mediation models to test the associations between MHT history, diabetes incidence, and late life cognitive performance. RESULTS 1,088/1,792 women (60.7%) reported a history of midlife MHT use and 220/1,792 (12.3%) reported a history of diabetes. MHT use history was associated with better late life immediate recall (but not delayed recall), as well as a reduced risk of diabetes with protracted time to onset. Causal mediation models suggest that the beneficial effect of midlife MHT use on late life immediate recall were at least partially mediated by diabetes risk. CONCLUSION Our data support a beneficial effect of MHT use on late life immediate recall (learning) that was partially mediated by protection against diabetes risk, supporting MHT use in midlife as protective against late life cognitive decline and adverse health outcomes.
Collapse
Affiliation(s)
- Victoria J. Williams
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Rebecca Koscik
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Kamil Sicinski
- Center for Demography of Health and Aging, University of Wisconsin at Madison, Madison, WI, USA
| | - Sterling C. Johnson
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Veterans Hospital, Madison, WI, USA
| | - Pamela Herd
- McCourt School of Public Policy, Georgetown University, Washington, DC, USA
| | - Sanjay Asthana
- Division of Geriatrics and Gerontology, Department of Medicine, University of Wisconsin at Madison, School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Veterans Hospital, Madison, WI, USA
| |
Collapse
|
23
|
Isoflavone-Enriched Soybean Leaves (Glycine Max) Alleviate Cognitive Impairment Induced by Ovariectomy and Modulate PI3K/Akt Signaling in the Hippocampus of C57BL6 Mice. Nutrients 2022; 14:nu14224753. [PMID: 36432439 PMCID: PMC9697522 DOI: 10.3390/nu14224753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/24/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022] Open
Abstract
(1) Background: The estrogen decline during perimenopause can induce various disorders, including cognitive impairment. Phytoestrogens, such as isoflavones, lignans, and coumestans, have been tried as a popular alternative to avoid the side effects of conventional hormone replacement therapy, but their exact mechanisms and risk are not fully elucidated. In this study, we investigated the effects of isoflavone-enriched soybean leaves (IESLs) on the cognitive impairment induced by ovariectomy in female mice. (2) Methods: Ovariectomy was performed at 9 weeks of age to mimic menopausal women, and the behavior tests for cognition were conducted 15 weeks after the first administration. IESLs were administered for 18 weeks. (3) Results: The present study showed the effects of IESLs on the cognitive function in the OVX (ovariectomized) mice. Ovariectomy markedly increased the body weight and fat accumulation in the liver and perirenal fat, but IESL treatment significantly inhibited them. In the behavioral tests, ovariectomy impaired cognitive functions, but administration of IESLs restored it. In addition, in the OVX mice, administration of IESLs restored decreased estrogen receptor (ER) β and PI3K/Akt expression in the hippocampus. (4) Conclusions: The positive effects of IESLs on cognitive functions may be closely related to the ER-mediated PI3/Akt signaling pathway in the hippocampus.
Collapse
|
24
|
Vigil P, Meléndez J, Petkovic G, Del Río JP. The importance of estradiol for body weight regulation in women. Front Endocrinol (Lausanne) 2022; 13:951186. [PMID: 36419765 PMCID: PMC9677105 DOI: 10.3389/fendo.2022.951186] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022] Open
Abstract
Obesity in women of reproductive age has a number of adverse metabolic effects, including Type II Diabetes (T2D), dyslipidemia, and cardiovascular disease. It is associated with increased menstrual irregularity, ovulatory dysfunction, development of insulin resistance and infertility. In women, estradiol is not only critical for reproductive function, but they also control food intake and energy expenditure. Food intake is known to change during the menstrual cycle in humans. This change in food intake is largely mediated by estradiol, which acts directly upon anorexigenic and orexigenic neurons, largely in the hypothalamus. Estradiol also acts indirectly with peripheral mediators such as glucagon like peptide-1 (GLP-1). Like estradiol, GLP-1 acts on receptors at the hypothalamus. This review describes the physiological and pathophysiological mechanisms governing the actions of estradiol during the menstrual cycle on food intake and energy expenditure and how estradiol acts with other weight-controlling molecules such as GLP-1. GLP-1 analogs have proven to be effective both to manage obesity and T2D in women. This review also highlights the relationship between steroid hormones and women's mental health. It explains how a decline or imbalance in estradiol levels affects insulin sensitivity in the brain. This can cause cerebral insulin resistance, which contributes to the development of conditions such as Parkinson's or Alzheimer's disease. The proper use of both estradiol and GLP-1 analogs can help to manage obesity and preserve an optimal mental health in women by reducing the mechanisms that trigger neurodegenerative disorders.
Collapse
Affiliation(s)
- Pilar Vigil
- Reproductive Health Research Institute (RHRI), Santiago, Chile
| | - Jaime Meléndez
- Reproductive Health Research Institute (RHRI), Santiago, Chile
| | - Grace Petkovic
- Arrowe Park Hospital, Department of Paediatrics, Wirral CH49 5PE, Merseyside, United Kingdom
| | - Juan Pablo Del Río
- Unidad de Psiquiatría Infantil y del Adolescente, Clínica Psiquiátrica Universitaria, Universidad de Chile, Santiago, Chile
- Millennium Nucleus to Improve the Mental Health of Adolescents and Youths, Millennium Science Initiative, Santiago, Chile
| |
Collapse
|
25
|
Nicotinic receptors promote susceptibility to social stress in female mice linked with neuroadaptations within VTA dopamine neurons. Neuropsychopharmacology 2022; 47:1587-1596. [PMID: 35459925 PMCID: PMC9283477 DOI: 10.1038/s41386-022-01314-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 12/14/2022]
Abstract
There are about twice as many women as men who experience depression during their lifetime. Although life circumstances and especially exposure to stressful situations constitute a major risk factor to develop depression, the underlying mechanisms have yet to be unraveled. We employed the chronic social defeat procedure to elicit depressive-like symptoms in females and ketamine to validate the model. We performed ex-vivo patch clamp recordings to assess cellular adaptations and used pharmacological agents to dissect these deregulations. Chronic social defeat exposure triggers a hyperactivity of VTA putative dopamine (DA) neurons in females susceptible to stress but not resilient ones. This hyperactivity was fully reversed by a single administration of ketamine. In virally-identified brain circuits of both susceptible and resilient females, we found a hypercholinergic tone to the VTA arising from the laterodorsal tegmentum. Application of puffs of nicotine revealed a decreased sensitivity of DA neurons in resilient mice when compared to naive or susceptible ones. The in vivo acute administration of the positive allosteric modulator for α7 nicotinic acetylcholine receptors (nAChRs) not only increased susceptibility to stress by enhancing activity of VTA DA neurons, but also triggered a switch in phenotype from resilient to susceptible. Our data unravel dysregulations of VTA DA neurons activity exclusively in females exhibiting depressive-like symptoms and identify VTA nAChRs as key molecular substrates that exacerbate susceptibility to stress.
Collapse
|
26
|
Debray A, Bherer L, Gagnon C, Bosquet L, Hay E, Bartlett AA, Gagnon D, Enea C. A Cross-Sectional Comparison of Arterial Stiffness and Cognitive Performances in Physically Active Late Pre- and Early Post-Menopausal Females. Brain Sci 2022; 12:brainsci12070901. [PMID: 35884708 PMCID: PMC9312988 DOI: 10.3390/brainsci12070901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 12/04/2022] Open
Abstract
Menopause accelerates increases in arterial stiffness and decreases cognitive performances. The objective of this study was to compare cognitive performances in physically active pre- and post-menopausal females and their relationship with arterial stiffness. We performed a cross-sectional comparison of blood pressure, carotid−femoral pulse wave velocity (cf-PWV) and cognitive performances between physically active late pre- and early post-menopausal females. Systolic (post-menopause—pre-menopause: +6 mmHg [95% CI −1; +13], p = 0.27; ŋ2 = 0.04) and diastolic (+6 mmHg [95% CI +2; +11], p = 0.06; ŋ2 = 0.12) blood pressures, and cf-PWV (+0.29 m/s [95% CI −1.03; 1.62], p = 0.48; ŋ2 = 0.02) did not differ between groups. Post-menopausal females performed as well as pre-menopausal females on tests evaluating executive functions, episodic memory and processing speed. Group differences were observed on the computerized working memory task. Post-menopausal females had lower accuracy (p = 0.02; ŋ2 = 0.25) but similar reaction time (p = 0.70; ŋ2 < 0.01). Moreover, this performance was inversely associated with the severity of menopausal symptoms (r = −0.38; p = 0.05). These results suggest that arterial stiffness and performance on tests assessing episodic memory and processing speed and executive functions assessing inhibition and switching abilities did not differ between physically active pre- and post-menopausal females. However, post-menopausal females had lower performance on a challenging condition of a working memory task, and this difference in working memory between groups cannot be explained by increased arterial stiffness.
Collapse
Affiliation(s)
- Amélie Debray
- Montreal Heart Institute, 5055 Rue Saint-Zotique E, Montreal, QC H1T 1N6, Canada; (A.D.); (L.B.); (C.G.); (A.-A.B.); (D.G.)
- School of Kinesiology and Exercise Science, Université de Montréal, 2100, boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
- Laboratory MOVE (UR20296), Faculté des Sciences du Sport, Université de Poitiers, Batiment C6, 8 allée Jean Monnet, TSA 31113, CEDEX 9, 86073 Poitiers, France; (L.B.); (E.H.)
| | - Louis Bherer
- Montreal Heart Institute, 5055 Rue Saint-Zotique E, Montreal, QC H1T 1N6, Canada; (A.D.); (L.B.); (C.G.); (A.-A.B.); (D.G.)
- Centre de Recherche de l’Institut Universitaire de Gériatrie de Montréal, 4565 Chemin Queen Mary, Montreal, QC H3W 1W5, Canada
- Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Christine Gagnon
- Montreal Heart Institute, 5055 Rue Saint-Zotique E, Montreal, QC H1T 1N6, Canada; (A.D.); (L.B.); (C.G.); (A.-A.B.); (D.G.)
- Centre de Recherche de l’Institut Universitaire de Gériatrie de Montréal, 4565 Chemin Queen Mary, Montreal, QC H3W 1W5, Canada
| | - Laurent Bosquet
- Laboratory MOVE (UR20296), Faculté des Sciences du Sport, Université de Poitiers, Batiment C6, 8 allée Jean Monnet, TSA 31113, CEDEX 9, 86073 Poitiers, France; (L.B.); (E.H.)
- Centre de Recherche de l’Institut Universitaire de Gériatrie de Montréal, 4565 Chemin Queen Mary, Montreal, QC H3W 1W5, Canada
| | - Eva Hay
- Laboratory MOVE (UR20296), Faculté des Sciences du Sport, Université de Poitiers, Batiment C6, 8 allée Jean Monnet, TSA 31113, CEDEX 9, 86073 Poitiers, France; (L.B.); (E.H.)
| | - Audrey-Ann Bartlett
- Montreal Heart Institute, 5055 Rue Saint-Zotique E, Montreal, QC H1T 1N6, Canada; (A.D.); (L.B.); (C.G.); (A.-A.B.); (D.G.)
- School of Kinesiology and Exercise Science, Université de Montréal, 2100, boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Daniel Gagnon
- Montreal Heart Institute, 5055 Rue Saint-Zotique E, Montreal, QC H1T 1N6, Canada; (A.D.); (L.B.); (C.G.); (A.-A.B.); (D.G.)
- School of Kinesiology and Exercise Science, Université de Montréal, 2100, boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
| | - Carina Enea
- Laboratory MOVE (UR20296), Faculté des Sciences du Sport, Université de Poitiers, Batiment C6, 8 allée Jean Monnet, TSA 31113, CEDEX 9, 86073 Poitiers, France; (L.B.); (E.H.)
- Correspondence: ; Tel.: +33-(0)5-49-45-30-00
| |
Collapse
|
27
|
Engler-Chiurazzi EB, Chastain WH, Citron KK, Lambert LE, Kikkeri DN, Shrestha SS. Estrogen, the Peripheral Immune System and Major Depression – A Reproductive Lifespan Perspective. Front Behav Neurosci 2022; 16:850623. [PMID: 35493954 PMCID: PMC9051447 DOI: 10.3389/fnbeh.2022.850623] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/17/2022] [Indexed: 12/01/2022] Open
Abstract
Major depression is a significant medical issue impacting millions of individuals worldwide. Identifying factors contributing to its manifestation has been a subject of intense investigation for decades and several targets have emerged including sex hormones and the immune system. Indeed, an extensive body of literature has demonstrated that sex hormones play a critical role in modulating brain function and impacting mental health, especially among female organisms. Emerging findings also indicate an inflammatory etiology of major depression, revealing new opportunities to supplement, or even supersede, currently available pharmacological interventions in some patient populations. Given the established sex differences in immunity and the profound impact of fluctuations of sex hormone levels on the immune system within the female, interrogating how the endocrine, nervous, and immune systems converge to impact women’s mental health is warranted. Here, we review the impacts of endogenous estrogens as well as exogenously administered estrogen-containing therapies on affect and immunity and discuss these observations in the context of distinct reproductive milestones across the female lifespan. A theoretical framework and important considerations for additional study in regards to mental health and major depression are provided.
Collapse
Affiliation(s)
- Elizabeth B. Engler-Chiurazzi
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane Brain Institute, Tulane University School of Medicine, New Orleans, LA, United States
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA, United States
- *Correspondence: Elizabeth B. Engler-Chiurazzi,
| | - Wesley H. Chastain
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane Brain Institute, Tulane University School of Medicine, New Orleans, LA, United States
| | - Kailen K. Citron
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane Brain Institute, Tulane University School of Medicine, New Orleans, LA, United States
| | - Lillian E. Lambert
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane Brain Institute, Tulane University School of Medicine, New Orleans, LA, United States
| | - Divya N. Kikkeri
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane Brain Institute, Tulane University School of Medicine, New Orleans, LA, United States
| | - Sharhana S. Shrestha
- Department of Neurosurgery, Clinical Neuroscience Research Center, Tulane Brain Institute, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
28
|
Estradiol and Estrogen-like Alternative Therapies in Use: The Importance of the Selective and Non-Classical Actions. Biomedicines 2022; 10:biomedicines10040861. [PMID: 35453610 PMCID: PMC9029610 DOI: 10.3390/biomedicines10040861] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/03/2022] [Accepted: 04/04/2022] [Indexed: 12/17/2022] Open
Abstract
Estrogen is one of the most important female sex hormones, and is indispensable for reproduction. However, its role is much wider. Among others, due to its neuroprotective effects, estrogen protects the brain against dementia and complications of traumatic injury. Previously, it was used mainly as a therapeutic option for influencing the menstrual cycle and treating menopausal symptoms. Unfortunately, hormone replacement therapy might be associated with detrimental side effects, such as increased risk of stroke and breast cancer, raising concerns about its safety. Thus, tissue-selective and non-classical estrogen analogues have become the focus of interest. Here, we review the current knowledge about estrogen effects in a broader sense, and the possibility of using selective estrogen-receptor modulators (SERMs), selective estrogen-receptor downregulators (SERDs), phytoestrogens, and activators of non-genomic estrogen-like signaling (ANGELS) molecules as treatment.
Collapse
|
29
|
Jett S, Malviya N, Schelbaum E, Jang G, Jahan E, Clancy K, Hristov H, Pahlajani S, Niotis K, Loeb-Zeitlin S, Havryliuk Y, Isaacson R, Brinton RD, Mosconi L. Endogenous and Exogenous Estrogen Exposures: How Women's Reproductive Health Can Drive Brain Aging and Inform Alzheimer's Prevention. Front Aging Neurosci 2022; 14:831807. [PMID: 35356299 PMCID: PMC8959926 DOI: 10.3389/fnagi.2022.831807] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/07/2022] [Indexed: 01/14/2023] Open
Abstract
After advanced age, female sex is the major risk factor for late-onset Alzheimer's disease (AD), the most common cause of dementia affecting over 24 million people worldwide. The prevalence of AD is higher in women than in men, with postmenopausal women accounting for over 60% of all those affected. While most research has focused on gender-combined risk, emerging data indicate sex and gender differences in AD pathophysiology, onset, and progression, which may help account for the higher prevalence in women. Notably, AD-related brain changes develop during a 10-20 year prodromal phase originating in midlife, thus proximate with the hormonal transitions of endocrine aging characteristic of the menopause transition in women. Preclinical evidence for neuroprotective effects of gonadal sex steroid hormones, especially 17β-estradiol, strongly argue for associations between female fertility, reproductive history, and AD risk. The level of gonadal hormones to which the female brain is exposed changes considerably across the lifespan, with relevance to AD risk. However, the neurobiological consequences of hormonal fluctuations, as well as that of hormone therapies, are yet to be fully understood. Epidemiological studies have yielded contrasting results of protective, deleterious and null effects of estrogen exposure on dementia risk. In contrast, brain imaging studies provide encouraging evidence for positive associations between greater cumulative lifetime estrogen exposure and lower AD risk in women, whereas estrogen deprivation is associated with negative consequences on brain structure, function, and biochemistry. Herein, we review the existing literature and evaluate the strength of observed associations between female-specific reproductive health factors and AD risk in women, with a focus on the role of endogenous and exogenous estrogen exposures as a key underlying mechanism. Chief among these variables are reproductive lifespan, menopause status, type of menopause (spontaneous vs. induced), number of pregnancies, and exposure to hormonal therapy, including hormonal contraceptives, hormonal therapy for menopause, and anti-estrogen treatment. As aging is the greatest risk factor for AD followed by female sex, understanding sex-specific biological pathways through which reproductive history modulates brain aging is crucial to inform preventative and therapeutic strategies for AD.
Collapse
Affiliation(s)
- Steven Jett
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Niharika Malviya
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Eva Schelbaum
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Grace Jang
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Eva Jahan
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Katherine Clancy
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Hollie Hristov
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Silky Pahlajani
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States
| | - Kellyann Niotis
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Susan Loeb-Zeitlin
- Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, United States
| | - Yelena Havryliuk
- Department of Obstetrics and Gynecology, Weill Cornell Medical College, New York, NY, United States
| | - Richard Isaacson
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| | - Roberta Diaz Brinton
- Department of Pharmacology, University of Arizona, Tucson, AZ, United States
- Department of Neurology, University of Arizona, Tucson, AZ, United States
| | - Lisa Mosconi
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
- Department of Radiology, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
30
|
Da Rocha RVO, Martins MIM, Antunes FTT, Martins MG, Klein AB, Corrêa DS, de Souza AH. Behavioral, Oxidative, and Biochemical Effects of Omega-3 on an Ovariectomized Rat Model of Menopause. J Menopausal Med 2022; 27:132-140. [PMID: 34989186 PMCID: PMC8738853 DOI: 10.6118/jmm.21016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/13/2021] [Accepted: 10/20/2021] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES Menopause induces changes in neuronal transmission, leading to anxiety and depression. Changes in the brain's glutamate levels cause psychological behavior in postmenopausal women. Omega-3 has been studied to improve some of these behaviors. METHODS Twenty-four female Wistar rats were divided into four groups: sham-operated treated with water (SO-W), sham-operated treated with omega-3 (SO-O), ovariectomized (OVX) treated with water (OVX-W), and bilateral OVX treated with omega-3 (OVX-O). These treatments were performed for 20 days via gavage, before and after surgery, totaling 40 days. RESULTS In the forced swimming, elevated plus-maze, and open field tests to assess behaviors, such as depression and anxiety, omega-3 improved these behaviors in both treated groups. The levels of thiobarbituric acid reactive substances (TBARS) in the brain were not different between the groups; however, there was a significant decrease in the catalase activity in the SO-O group compared with the SO-W group (P < 0.05). The glutamate level in the cerebrospinal fluid (CSF) was elevated in the SO-O group (P < 0.001) but not in the OVX-W or OVX-O groups. CONCLUSIONS These results bring novel data when related to the glutamatergic system in the SO-O group. This has suggested that the action mechanism of omega-3 was not dependent on glutamate levels in the CSF of the OVX group, but it played a regulatory role in the sham-operated animals. To confirm this, more studies are needed to explore this field when relating to the estrogen and glutamate receptor changes in specific brain regions.
Collapse
Affiliation(s)
| | | | | | - Marcia Gerhardt Martins
- Graduate Program in Genetic and Applied Toxicology, Lutheran University of Brazil (ULBRA), Canoas, Brazil
| | - Adriane Belló Klein
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Physiology Department, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Dione Silva Corrêa
- Graduate Program in Cellular and Molecular Biology, Lutheran University of Brazil (ULBRA), Canoas, Brazil
| | - Alessandra Hubner de Souza
- Graduate Program in Cellular and Molecular Biology, Lutheran University of Brazil (ULBRA), Canoas, Brazil.
| |
Collapse
|
31
|
Koszegi Z, Cheong RY. Targeting the non-classical estrogen pathway in neurodegenerative diseases and brain injury disorders. Front Endocrinol (Lausanne) 2022; 13:999236. [PMID: 36187099 PMCID: PMC9521328 DOI: 10.3389/fendo.2022.999236] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Estrogens can alter the biology of various tissues and organs, including the brain, and thus play an essential role in modulating homeostasis. Despite its traditional role in reproduction, it is now accepted that estrogen and its analogues can exert neuroprotective effects. Several studies have shown the beneficial effects of estrogen in ameliorating and delaying the progression of neurodegenerative diseases, including Alzheimer's and Parkinson's disease and various forms of brain injury disorders. While the classical effects of estrogen through intracellular receptors are more established, the impact of the non-classical pathway through receptors located at the plasma membrane as well as the rapid stimulation of intracellular signaling cascades are still under active research. Moreover, it has been suggested that the non-classical estrogen pathway plays a crucial role in neuroprotection in various brain areas. In this mini-review, we will discuss the use of compounds targeting the non-classical estrogen pathway in their potential use as treatment in neurodegenerative diseases and brain injury disorders.
Collapse
Affiliation(s)
- Zsombor Koszegi
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
| | - Rachel Y. Cheong
- Timeline Bioresearch AB, Medicon Village, Lund, Sweden
- *Correspondence: Rachel Y. Cheong,
| |
Collapse
|
32
|
Yu L, He M, Liu S, Dou X, Li L, Gu N, Li B, Liu Z, Wang G, Fan J. Fluorescent Egg White-Based Carbon Dots as a High-Sensitivity Iron Chelator for the Therapy of Nonalcoholic Fatty Liver Disease by Iron Overload in Zebrafish. ACS APPLIED MATERIALS & INTERFACES 2021; 13:54677-54689. [PMID: 34756030 DOI: 10.1021/acsami.1c14674] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Iron overload is the direct cause of many ferroptosis diseases, and it is essential to maintain iron homeostasis. In this paper, we report the Fe3+ chelation and therapy of the iron overload nonalcoholic fatty liver disease (NAFLD) by the fluorescent egg white-based carbon dots (EWCDs) obtained through the microwave-assisted pyrolysis method. As a high-sensitivity sensor, EWCDs show a high correlation between fluorescence emission and the concentration of Fe3+ (R2 = 0.993) in low concentration ranges of 0-25 μM. In vivo and in vitro, the EWCDs show characteristics of high biocompatibility and specific binding of Fe3+. As a novel type of the nano-iron-chelator, EWCDs can successfully attenuate the production of lethal reactive oxygen species. EWCDs not only alleviate the endoplasmic reticulum stress response but also regulate the NF-κB signaling pathway downstream of the Nrf2 signaling pathway. EWCDs prevent hepatocyte apoptosis, regulate fatty acid metabolism, and alleviate inflammation. Ultimately, they alleviate NAFLD induced by iron overload in zebrafish. This work may provide a new idea and method for the application of carbon dots in the field of disease detection and treatment.
Collapse
Affiliation(s)
- Lidong Yu
- School of Physics, Harbin Institute of Technology (HIT), Harbin 150080, P. R. China
| | - Mingyue He
- School of Life Science and Technology, HIT, Harbin 150080, P. R. China
| | - Sihan Liu
- School of Life Science and Technology, HIT, Harbin 150080, P. R. China
| | - Xinyue Dou
- School of Life Science and Technology, HIT, Harbin 150080, P. R. China
| | - Li Li
- School of Physics, Harbin Institute of Technology (HIT), Harbin 150080, P. R. China
- School of Life Science and Technology, HIT, Harbin 150080, P. R. China
| | - Ning Gu
- School of Life Science and Technology, HIT, Harbin 150080, P. R. China
| | - Bingsheng Li
- Key Laboratory of UV Light Emitting Materials and Technology of Ministry of Education, Northeast Normal University, Changchun 130024, P. R. China
| | - Zhiguo Liu
- School of Physics, Harbin Institute of Technology (HIT), Harbin 150080, P. R. China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Jianglin Fan
- Department of Molecular Pathology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi 409-3898, Japan
| |
Collapse
|
33
|
Wu CY, Lin YH, Hsieh HH, Lin JJ, Peng SL. Sex Differences in the Effect of Diabetes on Cerebral Glucose Metabolism. Biomedicines 2021; 9:1661. [PMID: 34829890 PMCID: PMC8615590 DOI: 10.3390/biomedicines9111661] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/05/2021] [Accepted: 11/08/2021] [Indexed: 12/25/2022] Open
Abstract
The neuroimaging literature indicates that brain structure and function both deteriorate with diabetes, but information on sexual dimorphism in diabetes-related brain alterations is limited. This study aimed to ascertain whether brain metabolism is influenced by sex in an animal model of diabetes. Eleven rats (male, n = 5; female, n = 6) received a single intraperitoneal injection of 70 mg/kg streptozotocin (STZ) to develop diabetes. Another 11 rats (male, n = 5; female, n = 6) received the same amount of solvent through a single intraperitoneal injection. Longitudinal positron emission tomography scans were used to assess cerebral glucose metabolism before and 4 weeks after STZ or solvent administration. Before STZ or solvent injections, there was no evidence of sexual dimorphism in cerebral metabolism (p > 0.05). Compared with healthy control animals, rats with diabetes had significantly decreased brain metabolism in all brain regions (all p < 0.05). In addition, female diabetic rats exhibited further reduction in cerebral metabolism, relative to male diabetic rats (p < 0.05). The results of this study may provide some biological evidence, supporting the existence of a sexual dimorphism in diabetes-related complications.
Collapse
Affiliation(s)
- Chun-Yi Wu
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei Branch, Taipei 112304, Taiwan; (C.-Y.W.); (H.-H.H.)
| | - Yu-Hsin Lin
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei Branch, Taipei 112304, Taiwan;
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404333, Taiwan
| | - Hsin-Hua Hsieh
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei Branch, Taipei 112304, Taiwan; (C.-Y.W.); (H.-H.H.)
| | - Jia-Jia Lin
- Center for Advanced Molecular Imaging and Translation, Chang Gung Memorial Hospital, Taoyuan 404333, Taiwan;
| | - Shin-Lei Peng
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung 404332, Taiwan
| |
Collapse
|
34
|
Hellman KM, Oladosu FA, Garrison EF, Roth GE, Dillane KE, Tu FF. Circulating sex steroids and bladder pain sensitivity in dysmenorrhea. Mol Pain 2021; 17:17448069211035217. [PMID: 34689649 DOI: 10.1177/17448069211035217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Although elevated estradiol levels facilitate chronic pelvic pain in animal models, it remains to be determined whether sex steroid levels are altered in a cross-section of women with chronic pelvic pain (CPP) and those at-risk for developing CPP. We sought to determine if sex steroid levels are increased in women with menstrual pain and whether those changes were more extreme in two groups of women with worsened pelvic pain profiles: a) dysmenorrhea plus evidence of bladder pain sensitivity and b) bladder pain syndrome. Serum samples were collected during the mid-luteal phase to measure estradiol, progesterone, testosterone, and sex hormone-binding globulin. We also compared quantitative sensory testing profiles to evaluate how sex steroid differences influence proposed pain sensitivity mechanisms. Women with combined dysmenorrhea and bladder sensitivity had higher estradiol concentrations than controls (487 [IQR 390 - 641] vs 404 [336 - 467] pmol/L, p = 0.042). Bladder pain syndrome participants had greater sex hormone-binding globulin than controls (83 [71 - 108] vs 55 [42 - 76 nmol/L; p = 0.027). Levels of pain sensitivity and mood were different across the groups, but the only significant relationship to sex steroids was that sex hormone-binding globulin was correlated to somatic symptoms (r = 0.26, p = 0.03). These findings show women potentially at-risk for CPP and women with diagnosed CPP exhibit altered circulating levels of sex steroids. Because these hormonal differences appear to be independent of mood or pain sensitivity, the role of sex steroids in the emergence of CPP may be via sensitization of visceral afferents.
Collapse
Affiliation(s)
- Kevin M Hellman
- Department of Obstetrics & Gynecology, Northshore University HealthSystem, Evanston, IL, USA.,Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Folabomi A Oladosu
- Department of Obstetrics & Gynecology, Northshore University HealthSystem, Evanston, IL, USA.,Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Ellen F Garrison
- Department of Obstetrics & Gynecology, Northshore University HealthSystem, Evanston, IL, USA
| | - Genevieve E Roth
- Department of Obstetrics & Gynecology, Northshore University HealthSystem, Evanston, IL, USA
| | - Katlyn E Dillane
- Department of Obstetrics & Gynecology, Northshore University HealthSystem, Evanston, IL, USA
| | - Frank F Tu
- Department of Obstetrics & Gynecology, Northshore University HealthSystem, Evanston, IL, USA.,Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
35
|
Zhao W, Hou Y, Song X, Wang L, Zhang F, Zhang H, Yu H, Zhou Y. Estrogen Deficiency Induces Mitochondrial Damage Prior to Emergence of Cognitive Deficits in a Postmenopausal Mouse Model. Front Aging Neurosci 2021; 13:713819. [PMID: 34335235 PMCID: PMC8319728 DOI: 10.3389/fnagi.2021.713819] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/28/2021] [Indexed: 01/04/2023] Open
Abstract
Background: Estrogen deficiency contributes to the development of Alzheimer's disease (AD) in menopausal women. In the current study, we examined the impact of estrogen deficiency on mitochondrial function and cognition using a postmenopausal mouse model. Methods: Bilateral ovariectomy was conducted in adult females C57BL/6J. Cognitive function was examined using the Morris water maze (MWM) test at 2 weeks, 1, 2, and 3 months after ovariectomy. Neurodegeneration was assessed using an immunofluorescence assay of microtubule-associated protein 2 (MAP2) in the hippocampus and immunoblotting against postsynaptic density-95 (PSD95). Mitochondrial function in the hippocampus was assessed using immunoblotting for NDUFB8, SDHB, UQCRC2, MTCO1, and ATP5A1. Mitochondrial biogenesis was examined using immunoblotting for PGC-1α, NRF1, and mtTFA. Mitochondrion fission was assessed with immunoblotting for Drp1, whereas mitochondrion fusion was analyzed with immunoblotting for OPA1 and Mfn2. Mitophagy was examined with immunoblotting for PINK1 and LC3B. Mice receiving sham surgery were used as controls. Results: Ovariectomy resulted in significant learning and memory deficits in the MWM test at 3 months, but not at any earlier time points. At 2 weeks after ovariectomy, levels of Drp1 phosphorylated at Ser637 decreased in the hippocampus. At 1 month after ovariectomy, hippocampal levels of NDUFB8, SDHB, PGC-1α, mtTFA, OPA1, and Mfn2 were significantly reduced. At 2 months after ovariectomy, hippocampal levels of MAP2, PSD95, MTCO1, NRF1, and Pink1 were also reduced. At 3 months, levels of LC3B-II were reduced. Conclusions: The cognitive decline associated with estrogen deficiency is preceded by mitochondrial dysfunction, abnormal mitochondrial biogenesis, irregular mitochondrial dynamics, and decreased mitophagy. Thus, mitochondrial damage may contribute to cognitive impairment associated with estrogen deficiency.
Collapse
Affiliation(s)
- Wei Zhao
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Yue Hou
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Xinxin Song
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Lei Wang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Fangfang Zhang
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Hanting Zhang
- Departments of Neuroscience and Behavioral Medicine and Psychiatry, Rockefeller Neurosciences Institute, West Virginia University Health Sciences Center, Morgantown, WV, United States
| | - Haiyang Yu
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Yanmeng Zhou
- Institute of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| |
Collapse
|
36
|
Androvičová R, Pfaus JG, Ovsepian SV. Estrogen pendulum in schizophrenia and Alzheimer's disease: Review of therapeutic benefits and outstanding questions. Neurosci Lett 2021; 759:136038. [PMID: 34116197 DOI: 10.1016/j.neulet.2021.136038] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/21/2021] [Accepted: 06/06/2021] [Indexed: 12/29/2022]
Abstract
Although produced largely in the periphery, gonadal steroids play a key role in regulating the development and functions of the central nervous system and have been implicated in several chronic neuropsychiatric disorders, with schizophrenia and Alzheimer's disease (AD) most prominent. Despite major differences in pathobiology and clinical manifestations, in both conditions, estrogen transpires primarily with protective effects, buffering the onset and progression of diseases at various levels. As a result, estrogen replacement therapy (ERT) emerges as one of the most widely discussed adjuvant interventions. In this review, we revisit evidence supporting the protective role of estrogen in schizophrenia and AD and consider putative cellular and molecular mechanisms. We explore the underlying functional processes relevant to the manifestation of these devastating conditions, with a focus on synaptic transmission and plasticity mechanisms. We discuss specific effects of estrogen deficit on neurotransmitter systems such as cholinergic, dopaminergic, serotoninergic, and glutamatergic. While the evidence from both, preclinical and clinical reports, in general, are supportive of the protective effects of estrogen from cognitive decline to synaptic pathology, numerous questions remain, calling for further research.
Collapse
Affiliation(s)
- Renáta Androvičová
- Department of Applied Neuroscience and Neuroimaging (RA) and Department of Experimental Neuroscience (SVO), National Institute of Mental Health, Topolová 748, 250 67 Klecany, Czech Republic.
| | - James G Pfaus
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa, Mexico
| | - Saak V Ovsepian
- Department of Applied Neuroscience and Neuroimaging (RA) and Department of Experimental Neuroscience (SVO), National Institute of Mental Health, Topolová 748, 250 67 Klecany, Czech Republic
| |
Collapse
|
37
|
Kim YJ, Soto M, Branigan GL, Rodgers K, Brinton RD. Association between menopausal hormone therapy and risk of neurodegenerative diseases: Implications for precision hormone therapy. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12174. [PMID: 34027024 PMCID: PMC8118114 DOI: 10.1002/trc2.12174] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/18/2021] [Accepted: 03/12/2021] [Indexed: 01/03/2023]
Abstract
INTRODUCTION The impact of menopausal hormone therapy (HT) on age-associated Alzheimer's and neurodegenerative diseases (NDDs) remains unresolved. To determine the effect of HT, formulation, type, and duration on risk of NDDs, a retrospective analysis was performed using a 10-year Humana claims dataset. METHODS Study population included women aged 45 years or older with or without claim records of HT medications. Patients diagnosed with NDDs including Alzheimer's disease (AD), Parkinson's disease (PD), dementia, multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS) were identified. Relative risk (RR) ratios and 95% confidence intervals (CI) for combined NDDs, or AD, PD, dementia, MS, and ALS were determined. Cumulative hazard ratios were determined to investigate the association between HT and NDDs at different age groups. RESULTS In 379,352 women with or without claim records of HT, use of HT was associated with significantly reduced risk for combined NDDs (RR 0.42, 95% CI 0.40-0.43, P < 0.001). Average follow-up time was 5.1 [2.3] years. Formulations containing natural steroids 17β-estradiol and/or progesterone were associated with greater reduction in NDD risk. Oral- HT users showed significantly reduced RRs (0.42, 0.41-0.44, P < 0.001) for combined NDDs compared to non-HT users. The RRs for transdermal-HT users were significantly decreased for all-cause dementia (0.73, 0.60-0.88, P = 0.001) and MS (0.55, 0.36-0.84, P = 0.005). Greatest reduction in risk of NDD, AD, and dementia emerged in patients aged 65 years or older. Further, the protective effect of long-term therapy (>1 year) on combined NDDs, AD, PD, and dementia was greater compared to short-term therapy (≤1 year). DISCUSSION HT was associated with reduced risk of all NDDs including AD and dementia, with greater duration of therapy and natural steroid formulations associated with greater efficacy. These findings advance precision HT to prevent NDDs including AD.
Collapse
Affiliation(s)
- Yu Jin Kim
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
| | - Maira Soto
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of PharmacologyUniversity of ArizonaTucsonArizonaUSA
| | - Gregory L Branigan
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of PharmacologyUniversity of ArizonaTucsonArizonaUSA
- College of MedicineMD‐PhD Training ProgramUniversity of ArizonaTucsonArizonaUSA
| | - Kathleen Rodgers
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of PharmacologyUniversity of ArizonaTucsonArizonaUSA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of PharmacologyUniversity of ArizonaTucsonArizonaUSA
- College of Medicine, Department of NeurologyUniversity of ArizonaTucsonArizonaUSA
| |
Collapse
|
38
|
Alonso A, Genzel L, Gomez A. Sex and Menstrual Phase Influences on Sleep and Memory. CURRENT SLEEP MEDICINE REPORTS 2021. [DOI: 10.1007/s40675-020-00201-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Abstract
Purposes of Review
This review highlights the effect of sex differences in sleep mediated memory consolidation and cognitive performance. In addition, the role of menstrual cycle and the fluctuating level of sexual hormones (mainly oestrogen and progesterone) are stressed.
Recent Findings
The literature indicates that sex hormones mediate and orchestrate the differences observed in performance of females in comparison with males in a variety of tasks and can also be related to how sleep benefits cognition. Although the exact mechanism of such influence is not clear, it most likely involves differential activation of brain areas, sensitivity to neuromodulators (mainly oestrogen), circadian regulation of sleep and temperature, as well as modification of strategies to solve tasks across the menstrual cycle.
Summary
With the evidence presented here, we hope to encourage researchers to develop appropriate paradigms to study the complex relationship between menstrual cycle, sleep (its regulation, architecture and electrophysiological hallmarks) and performance in memory and other cognitive tasks.
Collapse
|
39
|
Zeydan B, Lowe VJ, Tosakulwong N, Lesnick TG, Senjem ML, Jack CR, Fields JA, James TT, Gleason CE, Dowling NM, Miller VM, Kantarci K. Sleep quality and cortical amyloid-β deposition in postmenopausal women of the Kronos early estrogen prevention study. Neuroreport 2021; 32:326-331. [PMID: 33470769 PMCID: PMC7878341 DOI: 10.1097/wnr.0000000000001592] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Hormone therapy improves sleep in menopausal women and recent data suggest that transdermal 17β-estradiol may reduce the accumulation of cortical amyloid-β. However, how menopausal hormone therapies modify the associations of amyloid-β accumulation with sleep quality is not known. In this study, associations of sleep quality with cortical amyloid-β deposition and cognitive function were assessed in a subset of women who had participated in the Kronos early estrogen prevention study. It was a randomized, placebo-controlled trial in which recently menopausal women (age, 42-58; 5-36 months past menopause) were randomized to (1) oral conjugated equine estrogen (n = 19); (2) transdermal 17β-estradiol (tE2, n = 21); (3) placebo pills and patch (n = 32) for 4 years. Global sleep quality score was calculated using Pittsburgh sleep quality index, cortical amyloid-β deposition was measured with Pittsburgh compound-B positron emission tomography standard uptake value ratio and cognitive function was assessed in four cognitive domains 3 years after completion of trial treatments. Lower global sleep quality score (i.e., better sleep quality) correlated with lower cortical Pittsburgh compound-B standard uptake value ratio only in the tE2 group (r = 0.45, P = 0.047). Better global sleep quality also correlated with higher visual attention and executive function scores in the tE2 group (r = -0.54, P = 0.02) and in the oral conjugated equine estrogen group (r = -0.65, P = 0.005). Menopausal hormone therapies may influence the effects of sleep on cognitive function, specifically, visual attention and executive function. There also appears to be a complex relationship between sleep, menopausal hormone therapies, cortical amyloid-β accumulation and cognitive function, and tE2 formulation may modify the relationship between sleep and amyloid-β accumulation.
Collapse
Affiliation(s)
- Burcu Zeydan
- Department of Radiology, Mayo Clinic Rochester MN
- Department of Neurology, Mayo Clinic Rochester MN
| | - Val J. Lowe
- Department of Radiology, Mayo Clinic Rochester MN
| | | | | | - Matthew L. Senjem
- Department of Radiology, Mayo Clinic Rochester MN
- Department of Information Technology, Mayo Clinic Rochester MN
| | | | | | - Taryn T. James
- Division of Geriatrics, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI
- Wisconsin Alzheimer’s Disease Research Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI
| | - Carey E. Gleason
- Division of Geriatrics, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI
- Geriatric Research, Education and Clinical Center, William S. Middleton Memorial, Veterans Hospital, Madison, WI
| | - N. Maritza Dowling
- Department of Acute & Chronic Care, School of Nursing, Department of Epidemiology & Biostatistics, Milken Institute School of Public Health, The George Washington University, Washington, DC
| | - Virginia M. Miller
- Department of Physiology and Biomedical Engineering, Mayo Clinic Rochester MN
- Department of Surgery, Mayo Clinic Rochester MN
| | | |
Collapse
|
40
|
Effects of oral contraceptive pills on mood and magnetic resonance imaging measures of prefrontal cortical thickness. Mol Psychiatry 2021; 26:917-926. [PMID: 33420480 PMCID: PMC7914152 DOI: 10.1038/s41380-020-00990-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/23/2020] [Accepted: 12/07/2020] [Indexed: 01/29/2023]
Abstract
Gonadal hormones influence neuronal organization and plasticity. Yet the consequences of altering their concentrations by administering contraceptive agents, which are used by most reproductive-age women in the United States, are unclear. Cross-sectional studies have found both larger and smaller cortical regions alongside a variety of mood alterations in women who use oral contraceptive pills (OCPs) compared to naturally-cycling women. The goal of this study, therefore, was to determine whether there is an effect of OCPs on MRI measures of prefrontal cortical brain structure that may influence regulation of mood. We performed a double-blind, placebo-controlled, randomized crossover study comparing effects of OCPs (0.15 mg levonorgestrel + 0.30 μg ethinyl estradiol) vs placebo (N = 26) on MRI measures of prefrontal cortical thickness and on mood, as indicated by self-report on the Daily Record of Severity of Problems, which also includes one item related to somatic symptoms. MRI measures that reflect cortical thickness were smaller bilaterally in the pars triangularis and in the pars opercularis and frontal pole of the right hemisphere during the OCP arm vs. placebo. Only the effect in the right pars triangularis survived multiple comparisons correction. Right pars triangularis MRI measures of cortical thickness were not related to mood symptoms, but negatively correlated across conditions with severity of somatic symptoms on the DSRP. The somatic symptoms and MRI measures may be independently related to the actions of steroid hormones in OCPs, with OCPs simultaneously inducing both more effects on MRI measures of cortical thickness and somatic symptoms.
Collapse
|
41
|
Zhang Z, Park JW, Ahn IS, Diamante G, Sivakumar N, Arneson D, Yang X, van Veen JE, Correa SM. Estrogen receptor alpha in the brain mediates tamoxifen-induced changes in physiology in mice. eLife 2021; 10:63333. [PMID: 33647234 PMCID: PMC7924955 DOI: 10.7554/elife.63333] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 01/25/2021] [Indexed: 12/24/2022] Open
Abstract
Adjuvant tamoxifen therapy improves survival in breast cancer patients. Unfortunately, long-term treatment comes with side effects that impact health and quality of life, including hot flashes, changes in bone density, and fatigue. Partly due to a lack of proven animal models, the tissues and cells that mediate these negative side effects are unclear. Here, we show that mice undergoing tamoxifen treatment experience changes in temperature, bone, and movement. Single-cell RNA sequencing reveals that tamoxifen treatment induces widespread gene expression changes in the hypothalamus and preoptic area (hypothalamus-POA). These expression changes are dependent on estrogen receptor alpha (ERα), as conditional knockout of ERα in the hypothalamus-POA ablates or reverses tamoxifen-induced gene expression. Accordingly, ERα-deficient mice do not exhibit tamoxifen-induced changes in temperature, bone, or movement. These findings provide mechanistic insight into the effects of tamoxifen on the hypothalamus-POA and indicate that ERα mediates several physiological effects of tamoxifen treatment in mice.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, United States.,Laboratory of Neuroendocrinology of the Brain Research Institute, University of California Los Angeles, Los Angeles, United States
| | - Jae Whan Park
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, United States.,Laboratory of Neuroendocrinology of the Brain Research Institute, University of California Los Angeles, Los Angeles, United States
| | - In Sook Ahn
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, United States
| | - Graciel Diamante
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, United States
| | - Nilla Sivakumar
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, United States.,Laboratory of Neuroendocrinology of the Brain Research Institute, University of California Los Angeles, Los Angeles, United States
| | - Douglas Arneson
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, United States
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, United States
| | - J Edward van Veen
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, United States.,Laboratory of Neuroendocrinology of the Brain Research Institute, University of California Los Angeles, Los Angeles, United States
| | - Stephanie M Correa
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, United States.,Laboratory of Neuroendocrinology of the Brain Research Institute, University of California Los Angeles, Los Angeles, United States
| |
Collapse
|
42
|
Chou HT, Wu PY, Huang JC, Chen SC, Ho WY. Late Menarche, Not Reproductive Period, Is Associated with Poor Cognitive Function in Postmenopausal Women in Taiwan. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:2345. [PMID: 33673620 PMCID: PMC7967768 DOI: 10.3390/ijerph18052345] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/17/2021] [Accepted: 02/23/2021] [Indexed: 01/09/2023]
Abstract
Female sex hormones such as estrogen and progesterone play an important role in the regulation of a woman's body, including cognition and neurogenesis. However, the effects of age at menarche and reproductive period on cognitive function are still controversial. The aim of this study was to investigate the relationships between age at menarche and reproductive period with cognitive impairment. Data were obtained from the Taiwan Biobank. Cognitive function was assessed using the Mini Mental State Examination (MMSE) and its five subdomains. Multivariable linear regression analysis revealed that an old age at menarche (per one year; coefficient β, -0.189; p = 0.020) was significantly associated with a low total MMSE score, whereas reproductive period (p = 0.733) was not significantly associated with total MMSE score. Furthermore, an old age at menarche was significantly associated with low MMSE G2 (registration) (per one year; coefficient β, -0.022; p = 0.035) and G5 (language, construction and obey) scores (per one year; coefficient β, -0.054; p = 0.047). However, age at menarche was not significantly associated with MMSE G1 (orientation), G3 (attention and calculation) and G4 (recall) scores. In addition, reproductive period was not significantly associated with any MMSE subscores. Late menarche was associated with poor cognitive function, including low total MMSE score and low MMSE G2 and G5 scores. However, reproductive period was not associated with cognitive function in postmenopausal women.
Collapse
Affiliation(s)
- Hung-Tse Chou
- Department of General Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan;
| | - Pei-Yu Wu
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan; (P.-Y.W.); (J.-C.H.); (S.-C.C.)
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Jiun-Chi Huang
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan; (P.-Y.W.); (J.-C.H.); (S.-C.C.)
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Szu-Chia Chen
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan; (P.-Y.W.); (J.-C.H.); (S.-C.C.)
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Wan-Yi Ho
- Department of Anatomy, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
43
|
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
|
44
|
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
|
45
|
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: 4.0] [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
|
46
|
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: 3.0] [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
|
47
|
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: 4.0] [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
|
48
|
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: 29] [Impact Index Per Article: 7.3] [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
|
49
|
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: 0.8] [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
|
50
|
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: 2.5] [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
|