101
|
Nagasaka Y, Wepler M, Thoonen R, Sips PY, Allen K, Graw JA, Yao V, Burns SM, Muenster S, Brouckaert P, Miller K, Solt K, Buys ES, Ichinose F, Zapol WM. Sensitivity to Sevoflurane anesthesia is decreased in mice with a congenital deletion of Guanylyl Cyclase-1 alpha. BMC Anesthesiol 2017; 17:76. [PMID: 28615047 PMCID: PMC5471676 DOI: 10.1186/s12871-017-0368-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 05/31/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Volatile anesthetics increase levels of the neurotransmitter nitric oxide (NO) and the secondary messenger molecule cyclic guanosine monophosphate (cGMP) in the brain. NO activates the enzyme guanylyl cyclase (GC) to produce cGMP. We hypothesized that the NO-GC-cGMP pathway contributes to anesthesia-induced unconsciousness. METHODS Sevoflurane-induced loss and return of righting reflex (LORR and RORR, respectively) were studied in wild-type mice (WT) and in mice congenitally deficient in the GC-1α subunit (GC-1-/- mice). Spatial distributions of GC-1α and the GC-2α subunit in the brain were visualized by in situ hybridization. Brain cGMP levels were measured in WT and GC-1-/- mice after inhaling oxygen with or without 1.2% sevoflurane for 20 min. RESULTS Higher concentrations of sevoflurane were required to induce LORR in GC-1-/- mice than in WT mice (1.5 ± 0.1 vs. 1.1 ± 0.2%, respectively, n = 14 and 14, P < 0.0001). Similarly, RORR occurred at higher concentrations of sevoflurane in GC-1-/- mice than in WT mice (1.0 ± 0.1 vs. 0.8 ± 0.1%, respectively, n = 14 and 14, P < 0.0001). Abundant GC-1α and GC-2α mRNA expression was detected in the cerebral cortex, medial habenula, hippocampus, and cerebellum. Inhaling 1.2% sevoflurane for 20 min increased cGMP levels in the brains of WT mice from 2.6 ± 2.0 to 5.5 ± 3.7 pmol/mg protein (n = 13 and 10, respectively, P = 0.0355) but not in GC-1-/- mice. CONCLUSION Congenital deficiency of GC-1α abolished the ability of sevoflurane anesthesia to increase cGMP levels in the whole brain, and increased the concentration of sevoflurane required to induce LORR. Impaired NO-cGMP signaling raises the threshold for producing sevoflurane-induced unconsciousness in mice.
Collapse
Affiliation(s)
- Yasuko Nagasaka
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Martin Wepler
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Robrecht Thoonen
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Patrick Y Sips
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Kaitlin Allen
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jan A Graw
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vincent Yao
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sara M Burns
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium and Inflammation Research Center, VIB, Ghent, Belgium
| | - Stefan Muenster
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Peter Brouckaert
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Keith Miller
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ken Solt
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Emmanuel S Buys
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Fumito Ichinose
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Warren M Zapol
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
102
|
Zárate S, Astiz M, Magnani N, Imsen M, Merino F, Álvarez S, Reinés A, Seilicovich A. Hormone deprivation alters mitochondrial function and lipid profile in the hippocampus. J Endocrinol 2017; 233:1-14. [PMID: 28130408 DOI: 10.1530/joe-16-0451] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 01/27/2017] [Indexed: 12/22/2022]
Abstract
Mitochondrial dysfunction is a common hallmark in aging. In the female, reproductive senescence is characterized by loss of ovarian hormones, many of whose neuroprotective effects converge upon mitochondria. The functional integrity of mitochondria is dependent on membrane fatty acid and phospholipid composition, which are also affected during aging. The effect of long-term ovarian hormone deprivation upon mitochondrial function and its putative association with changes in mitochondrial membrane lipid profile in the hippocampus, an area primarily affected during aging and highly responsive to ovarian hormones, is unknown. To this aim, Wistar adult female rats were ovariectomized or sham-operated. Twelve weeks later, different parameters of mitochondrial function (O2 uptake, ATP production, membrane potential and respiratory complex activities) as well as membrane phospholipid content and composition were evaluated in hippocampal mitochondria. Chronic ovariectomy reduced mitochondrial O2 uptake and ATP production rates and induced membrane depolarization during active respiration without altering the activity of respiratory complexes. Mitochondrial membrane lipid profile showed no changes in cholesterol levels but higher levels of unsaturated fatty acids and a higher peroxidizability index in mitochondria from ovariectomized rats. Interestingly, ovariectomy also reduced cardiolipin content and altered cardiolipin fatty acid profile leading to a lower peroxidizability index. In conclusion, chronic ovarian hormone deprivation induces mitochondrial dysfunction and changes in the mitochondrial membrane lipid profile comparable to an aging phenotype. Our study provides insights into ovarian hormone loss-induced early lipidomic changes with bioenergetic deficits in the hippocampus that may contribute to the increased risk of Alzheimer's disease and other age-associated disorders observed in postmenopause.
Collapse
Affiliation(s)
- Sandra Zárate
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET)Buenos Aires, Argentina
- Departamento de HistologíaEmbriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariana Astiz
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP, UNLP-CONICET) and Cátedra de Bioquímica y Biología MolecularFacultad de Medicina, Universidad Nacional de La Plata, La Plata, Argentina
| | - Natalia Magnani
- Instituto de Bioquímica y Medicina Molecular (IBIMOL, UBA-CONICET)Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mercedes Imsen
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET)Buenos Aires, Argentina
| | - Florencia Merino
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET)Buenos Aires, Argentina
| | - Silvia Álvarez
- Instituto de Bioquímica y Medicina Molecular (IBIMOL, UBA-CONICET)Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Analía Reinés
- Instituto de Biología Celular y Neurociencias 'Prof. E. De Robertis' (IBCN, UBA-CONICET)Facultad de Medicina and Cátedra de Farmacología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Adriana Seilicovich
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET)Buenos Aires, Argentina
- Departamento de HistologíaEmbriología, Biología Celular y Genética, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
103
|
Chen X, Tao L, Li J, Wu J, Zhu C, Yu F, Zhang L, Zhang J, Qiu B, Yu Y, Wang K, Wang K. The Working Memory and Dorsolateral Prefrontal-Hippocampal Functional Connectivity Changes in Long-Term Survival Breast Cancer Patients Treated with Tamoxifen. Int J Neuropsychopharmacol 2017; 20:374-382. [PMID: 28177081 PMCID: PMC5417059 DOI: 10.1093/ijnp/pyx008] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 02/02/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Tamoxifen is the most widely used drug for treating patients with estrogen receptor-sensitive breast cancer. There is evidence that breast cancer patients treated with tamoxifen exhibit cognitive dysfunction. However, the underlying neural mechanism remains unclear. The present study aimed to investigate the neural mechanisms underlying working memory deficits in combination with functional connectivity changes in premenopausal women with breast cancer who received long-term tamoxifen treatment. METHODS A total of 31 premenopausal women with breast cancer who received tamoxifen and 32 matched healthy control participants were included. The participants completed n-back tasks and underwent resting-state functional magnetic resonance imaging, which measure working memory performance and brain functional connectivity, respectively. A seed-based functional connectivity analysis within the whole brain was conducted, for which the dorsolateral prefrontal cortex was chosen as the seed region. RESULTS Our results indicated that the tamoxifen group had significant deficits in working memory and general executive function performance and significantly lower functional connectivity of the right dorsolateral prefrontal cortex with the right hippocampus compared with the healthy controls. There were no significant changes in functional connectivity in the left dorsolateral prefrontal cortex within the whole brain between the tamoxifen group and healthy controls. Moreover, significant correlations were found in the tamoxifen group between the functional connectivity strength of the dorsolateral prefrontal cortex with the right hippocampus and decreased working memory performance. CONCLUSION This study demonstrates that the prefrontal cortex and hippocampus may be affected by tamoxifen treatment, supporting an antagonistic role of tamoxifen in the long-term treatment of breast cancer patients.
Collapse
Affiliation(s)
- Xingui Chen
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Chen and Wang and Mrs Wu); Collaborative Innovation Centre of Neuropsychiatric Disorders and Mental Health, Anhui Province, China (Drs Chen, Zhu, Yu, L. Zhang, and Wang); Center for Biomedical Engineering, University of Science and Technology of China, Hefei, China (Dr Qiu and Mr He); Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Tao and Yu); Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (Mrs Li); Department of Medical Psychology, Anhui Medical University, Hefei, China (Drs Zhu, Yu, L. Zhang, and Wang); Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Dr J. Zhang)
| | - Longxiang Tao
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Chen and Wang and Mrs Wu); Collaborative Innovation Centre of Neuropsychiatric Disorders and Mental Health, Anhui Province, China (Drs Chen, Zhu, Yu, L. Zhang, and Wang); Center for Biomedical Engineering, University of Science and Technology of China, Hefei, China (Dr Qiu and Mr He); Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Tao and Yu); Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (Mrs Li); Department of Medical Psychology, Anhui Medical University, Hefei, China (Drs Zhu, Yu, L. Zhang, and Wang); Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Dr J. Zhang)
| | - Jingjing Li
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Chen and Wang and Mrs Wu); Collaborative Innovation Centre of Neuropsychiatric Disorders and Mental Health, Anhui Province, China (Drs Chen, Zhu, Yu, L. Zhang, and Wang); Center for Biomedical Engineering, University of Science and Technology of China, Hefei, China (Dr Qiu and Mr He); Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Tao and Yu); Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (Mrs Li); Department of Medical Psychology, Anhui Medical University, Hefei, China (Drs Zhu, Yu, L. Zhang, and Wang); Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Dr J. Zhang)
| | - Jiaonan Wu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Chen and Wang and Mrs Wu); Collaborative Innovation Centre of Neuropsychiatric Disorders and Mental Health, Anhui Province, China (Drs Chen, Zhu, Yu, L. Zhang, and Wang); Center for Biomedical Engineering, University of Science and Technology of China, Hefei, China (Dr Qiu and Mr He); Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Tao and Yu); Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (Mrs Li); Department of Medical Psychology, Anhui Medical University, Hefei, China (Drs Zhu, Yu, L. Zhang, and Wang); Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Dr J. Zhang)
| | - Chunyan Zhu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Chen and Wang and Mrs Wu); Collaborative Innovation Centre of Neuropsychiatric Disorders and Mental Health, Anhui Province, China (Drs Chen, Zhu, Yu, L. Zhang, and Wang); Center for Biomedical Engineering, University of Science and Technology of China, Hefei, China (Dr Qiu and Mr He); Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Tao and Yu); Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (Mrs Li); Department of Medical Psychology, Anhui Medical University, Hefei, China (Drs Zhu, Yu, L. Zhang, and Wang); Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Dr J. Zhang)
| | - Fengqiong Yu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Chen and Wang and Mrs Wu); Collaborative Innovation Centre of Neuropsychiatric Disorders and Mental Health, Anhui Province, China (Drs Chen, Zhu, Yu, L. Zhang, and Wang); Center for Biomedical Engineering, University of Science and Technology of China, Hefei, China (Dr Qiu and Mr He); Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Tao and Yu); Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (Mrs Li); Department of Medical Psychology, Anhui Medical University, Hefei, China (Drs Zhu, Yu, L. Zhang, and Wang); Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Dr J. Zhang)
| | - Lei Zhang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Chen and Wang and Mrs Wu); Collaborative Innovation Centre of Neuropsychiatric Disorders and Mental Health, Anhui Province, China (Drs Chen, Zhu, Yu, L. Zhang, and Wang); Center for Biomedical Engineering, University of Science and Technology of China, Hefei, China (Dr Qiu and Mr He); Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Tao and Yu); Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (Mrs Li); Department of Medical Psychology, Anhui Medical University, Hefei, China (Drs Zhu, Yu, L. Zhang, and Wang); Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Dr J. Zhang)
| | - Jingjie Zhang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Chen and Wang and Mrs Wu); Collaborative Innovation Centre of Neuropsychiatric Disorders and Mental Health, Anhui Province, China (Drs Chen, Zhu, Yu, L. Zhang, and Wang); Center for Biomedical Engineering, University of Science and Technology of China, Hefei, China (Dr Qiu and Mr He); Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Tao and Yu); Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (Mrs Li); Department of Medical Psychology, Anhui Medical University, Hefei, China (Drs Zhu, Yu, L. Zhang, and Wang); Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Dr J. Zhang)
| | - Bensheng Qiu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Chen and Wang and Mrs Wu); Collaborative Innovation Centre of Neuropsychiatric Disorders and Mental Health, Anhui Province, China (Drs Chen, Zhu, Yu, L. Zhang, and Wang); Center for Biomedical Engineering, University of Science and Technology of China, Hefei, China (Dr Qiu and Mr He); Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Tao and Yu); Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (Mrs Li); Department of Medical Psychology, Anhui Medical University, Hefei, China (Drs Zhu, Yu, L. Zhang, and Wang); Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Dr J. Zhang)
| | - Yongqiang Yu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Chen and Wang and Mrs Wu); Collaborative Innovation Centre of Neuropsychiatric Disorders and Mental Health, Anhui Province, China (Drs Chen, Zhu, Yu, L. Zhang, and Wang); Center for Biomedical Engineering, University of Science and Technology of China, Hefei, China (Dr Qiu and Mr He); Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Tao and Yu); Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (Mrs Li); Department of Medical Psychology, Anhui Medical University, Hefei, China (Drs Zhu, Yu, L. Zhang, and Wang); Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Dr J. Zhang)
| | - Kai Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Chen and Wang and Mrs Wu); Collaborative Innovation Centre of Neuropsychiatric Disorders and Mental Health, Anhui Province, China (Drs Chen, Zhu, Yu, L. Zhang, and Wang); Center for Biomedical Engineering, University of Science and Technology of China, Hefei, China (Dr Qiu and Mr He); Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Drs Tao and Yu); Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China (Mrs Li); Department of Medical Psychology, Anhui Medical University, Hefei, China (Drs Zhu, Yu, L. Zhang, and Wang); Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China (Dr J. Zhang)
| | - Kai Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Collaborative Innovation Centre of Neuropsychiatric Disorders and Mental Health, Anhui Province, China.,Department of Medical Psychology, Anhui Medical University, Hefei, China
| |
Collapse
|
104
|
Todorova V, Blokland A. Mitochondria and Synaptic Plasticity in the Mature and Aging Nervous System. Curr Neuropharmacol 2017; 15:166-173. [PMID: 27075203 PMCID: PMC5327446 DOI: 10.2174/1570159x14666160414111821] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 03/23/2016] [Accepted: 04/11/2016] [Indexed: 01/11/2023] Open
Abstract
Synaptic plasticity in the adult brain is believed to represent the cellular mechanisms of learning and memory. Mitochondria are involved in the regulation of the complex processes of synaptic plasticity. This paper reviews the current knowledge on the regulatory roles of mitochondria in the function and plasticity of synapses and the implications of mitochondrial dysfunctions in synaptic transmission. First, the importance of mitochondrial distribution and motility for maintenance and strengthening of dendritic spines, but also for new spines/synapses formation is presented. Secondly, the major mitochondrial functions as energy supplier and calcium buffer organelles are considered as possible explanation for their essential and regulatory roles in neuronal plasticity processes. Thirdly, the effects of synaptic potentiation on mitochondrial gene expression are discussed. And finally, the relation between age-related alterations in synaptic plasticity and mitochondrial dysfunctions is considered. It appears that memory loss and neurodegeneration during aging are related to mitochondrial (dys)function. Although, it is clear that mitochondria are essential for synaptic plasticity, further studies are indicated to scrutinize the intracellular and molecular processes that regulate the functions of mitochondria in synaptic plasticity.
Collapse
Affiliation(s)
- Vyara Todorova
- Institute II for Anatomy, Medical Faculty, University of Cologne, Cologne, Germany
| | | |
Collapse
|
105
|
Effect on Cognition of Estroprogestins Combined with Interferon Beta in Multiple Sclerosis: Analysis of Secondary Outcomes from a Randomised Controlled Trial. CNS Drugs 2017; 31:161-168. [PMID: 27995531 DOI: 10.1007/s40263-016-0401-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Cognitive impairment is a disabling symptom in multiple sclerosis (MS). While its management remains challenging, beneficial effects on cognition of interferon beta (IFN-β) have been reported and a positive effect from estroprogestins has been hypothesised, suggesting that the combination of the two medications in women with MS could offer a promising treatment strategy. OBJECTIVES We investigated whether a combination of estroprogestins and IFN-β can improve cognition in women with MS. METHODS Women with relapsing-remitting (RR) MS were randomly assigned (1:1:1) to receive subcutaneous IFN-β-1a (Rebif®, Merck Serono, Geneva, Switzerland) 44 mcg three times a week (tiw) (group 1), subcutaneous IFN-β-1a 44 mcg tiw plus ethinyl estradiol 20 mcg and desogestrel 150 mcg (Mercilon®, MSD Italia SRL, Rome, Italy) (group 2) or subcutaneous IFN-β-1a 44 mcg tiw plus ethinyl estradiol 40 mcg and desogestrel 125 mcg (Gracial®, Organon Italia S.p.A., Rome, Italy) (group 3) in a randomised controlled trial, for which we report the analysis of secondary outcomes. At baseline and at 24 months, all patients underwent magnetic resonance imaging (MRI) and a comprehensive cognitive assessment, including Rao's Brief Repeatable Battery (RBRB) and questionnaires for depression, fatigue and quality of life. Failure in at least two of the RBRB tests defined 'cognitive impairment'. RESULTS At baseline, there was no difference in the proportion of cognitively impaired patients. At month 24, the proportion of patients with cognitive impairment was lower in group 3 (34.8%) than in group 1 (47.6%) (p = 0.03). The risk of developing cognitive impairment over 24 months was lower in group 3 (p = 0.02). Mood and fatigue scores were comparable across the groups over time at both time points. However, at month 24, group 3 showed worsening on the sexual function subscale of the 54-item MS quality-of-life questionnaire (p = 0.03). CONCLUSIONS This study suggests that the combination of high-dose estroprogestins and IFN-β may have positive effects on cognition. However, the effect of this treatment on sexual function requires caution to be exercised. Protocol Number NCT00151801, registered in ClinicalTrials.gov.
Collapse
|
106
|
Alexander A, Irving AJ, Harvey J. Emerging roles for the novel estrogen-sensing receptor GPER1 in the CNS. Neuropharmacology 2017; 113:652-660. [DOI: 10.1016/j.neuropharm.2016.07.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 07/01/2016] [Accepted: 07/04/2016] [Indexed: 02/06/2023]
|
107
|
Lin KA, Rundel C, Doraiswamy PM. Serum SHBG Levels are not Associated with Longitudinal Cognitive Decline in Mild Cognitive Impairment. J Alzheimers Dis 2016; 55:1123-1130. [DOI: 10.3233/jad-160513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Katherine Amy Lin
- Department of Psychiatry, Duke University Medical Center, Durham, NC, USA
- Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| | - Colin Rundel
- Department of Statistical Science, Duke University, Durham, NC, USA
| | - P. Murali Doraiswamy
- Department of Psychiatry, Duke University Medical Center, Durham, NC, USA
- Duke Institute for Brain Sciences, Duke University, Durham, NC, USA
| | | |
Collapse
|
108
|
Wang X, Huang Y, Yuan S, Tamadon A, Ma S, Feng Y. The Role of Hippocampal Estradiol Receptor- α in a Perimenopausal Affective Disorders-Like Rat Model and Attenuating of Anxiety by Electroacupuncture. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2016; 2016:4958312. [PMID: 28044085 PMCID: PMC5156811 DOI: 10.1155/2016/4958312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 10/16/2016] [Accepted: 10/25/2016] [Indexed: 02/07/2023]
Abstract
Hormone replacement therapy is the principal treatment for perimenopausal affective disorders which can cause severe side effects. The present study compared the effects of electroacupuncture (EA) and estradiol treatment on perimenopausal affective disorders at the behavioral and cellular levels. In this randomized experimental in vivo study, adult female rats were divided into intact, ovariectomy, chronic unpredictable stress (CUS), and ovariectomy and CUS combination groups. After week 6, all groups were subdivided to three subgroups of control, EA, and estradiol treatment. The behavioral parameters in the open field and the elevated plus maze tests were assessed before and after treatments. Alterations of serum steroid hormones and changes of estradiol receptor-α (ER-α) immunofluorescence neurons in the hippocampus sections were evaluated. EA treatment caused more antianxiety effects than estradiol treatment in CUS group (P < 0.05). Notably, estradiol and EA treatments had better significant behavioral effects when the models were not estrogen-deficient. Importantly, within each group, compared to the control group, the numbers of ER-α-positive neurons were significantly larger in EA subgroups. Therefore, EA had antianxiety effects on perimenopausal affective disorders caused by CUS but not by estrogen deficiency and upregulation of hippocampus ER-α neurons may contribute to its mechanism of action.
Collapse
Affiliation(s)
- Xun Wang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, China
- 2008 Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yongheng Huang
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, China
- 2008 Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Shiwen Yuan
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, China
- 2008 Clinical Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Amin Tamadon
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, China
| | - Shulan Ma
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, China
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Institute of Acupuncture Research (WHO Collaborating Center for Traditional Medicine), Institutes of Brain Science, Brain Science Collaborative Innovation Center, Fudan University, Shanghai 200032, China
| |
Collapse
|
109
|
Karim R, Dang H, Henderson VW, Hodis HN, St John J, Brinton RD, Mack WJ. Effect of Reproductive History and Exogenous Hormone Use on Cognitive Function in Mid- and Late Life. J Am Geriatr Soc 2016; 64:2448-2456. [PMID: 27996108 DOI: 10.1111/jgs.14658] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES To investigate the association between reproductive history indicators of hormonal exposure, including reproductive period, pregnancy, and use of hormonal contraceptives, and mid- and late-life cognition in postmenopausal women. DESIGN Analysis of baseline data from two randomized clinical trials: the Women's Isoflavone Soy Health and the Early vs Late Intervention Trial of Estradiol. SETTING University academic research center. PARTICIPANTS Naturally menopausal women (N = 830). MEASUREMENTS Participants were uniformly evaluated using a cognitive battery and a structured reproductive history questionnaire. Outcomes were composite scores for verbal episodic memory, executive function, and global cognition. Reproductive variables included ages at pregnancies, menarche, and menopause; reproductive period; number of pregnancies; and use of hormones for contraception and menopausal symptoms. Multivariable linear regression was used to evaluate associations between cognitive scores (dependent variable) and reproductive factors (independent variables), adjusting for age, race and ethnicity, income, and education. RESULTS On multivariable modeling, age at menarche of 13 and older was inversely associated with global cognition (P = .05). Last pregnancy after age 35 was positively associated with verbal memory (P = .03). Use of hormonal contraceptives was positively associated with global cognition (P trend = .04), and verbal memory (P trend = .007). The association between hormonal contraceptive use and verbal memory and executive function was strongest for more than 10 years of use. Reproductive period was positively associated with global cognition (P = .04) and executive function (P = .04). CONCLUSION In this sample of healthy postmenopausal women, reproductive life events related to sex hormones, including earlier age at menarche, later age at last pregnancy, longer reproductive period, and use of oral contraceptives are positively related to aspects of cognition in later life.
Collapse
Affiliation(s)
- Roksana Karim
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California.,Atherosclerosis Research Unit, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Ha Dang
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Victor W Henderson
- Division of Epidemiology, Department of Health Research and Policy, Stanford University, Stanford, California.,Department of Neurology and Neurosciences, Stanford University, Stanford, California
| | - Howard N Hodis
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California.,Atherosclerosis Research Unit, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California.,Department of Pharmacology and Pharmaceutical Science, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Jan St John
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California.,Atherosclerosis Research Unit, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Roberta D Brinton
- Department of Pharmacology and Pharmaceutical Science, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Wendy J Mack
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California.,Atherosclerosis Research Unit, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
110
|
Wang Y, Brinton RD. Triad of Risk for Late Onset Alzheimer's: Mitochondrial Haplotype, APOE Genotype and Chromosomal Sex. Front Aging Neurosci 2016; 8:232. [PMID: 27757081 PMCID: PMC5047907 DOI: 10.3389/fnagi.2016.00232] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 09/20/2016] [Indexed: 01/02/2023] Open
Abstract
Brain is the most energetically demanding organ of the body, and is thus vulnerable to even modest decline in ATP generation. Multiple neurodegenerative diseases are associated with decline in mitochondrial function, e.g., Alzheimer’s, Parkinson’s, multiple sclerosis and multiple neuropathies. Genetic variances in the mitochondrial genome can modify bioenergetic and respiratory phenotypes, at both the cellular and system biology levels. Mitochondrial haplotype can be a key driver of mitochondrial efficiency. Herein, we focus on the association between mitochondrial haplotype and risk of late onset Alzheimer’s disease (LOAD). Evidence for the association of mitochondrial genetic variances/haplotypes and the risk of developing LOAD are explored and discussed. Further, we provide a conceptual framework that suggests an interaction between mitochondrial haplotypes and two demonstrated risk factors for Alzheimer’s disease (AD), apolipoprotein E (APOE) genotype and chromosomal sex. We posit herein that mitochondrial haplotype, and hence respiratory capacity, plays a key role in determining risk of LOAD and other age-associated neurodegenerative diseases. Further, therapeutic design and targeting that involve mitochondrial haplotype would advance precision medicine for AD and other age related neurodegenerative diseases.
Collapse
Affiliation(s)
- Yiwei Wang
- Department of Clinical Pharmacy, School of Pharmacy, University of Southern California Los Angeles, CA, USA
| | - Roberta D Brinton
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California Los Angeles, CA, USA
| |
Collapse
|
111
|
Impact of Sex and Menopausal Status on Episodic Memory Circuitry in Early Midlife. J Neurosci 2016; 36:10163-73. [PMID: 27683911 DOI: 10.1523/jneurosci.0951-16.2016] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 08/08/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Cognitive neuroscience of aging studies traditionally target participants age 65 and older. However, epidemiological surveys show that many women report increased forgetfulness earlier in the aging process, as they transition to menopause. In this population-based fMRI study, we stepped back by over a decade to characterize the changes in memory circuitry that occur in early midlife, as a function of sex and women's reproductive stage. Participants (N = 200; age range, 45-55) performed a verbal encoding task during fMRI scanning. Reproductive histories and serologic evaluations were used to determine menopausal status. Results revealed a pronounced impact of reproductive stage on task-evoked hippocampal responses, despite minimal difference in chronological age. Next, we examined the impact of sex and reproductive stage on functional connectivity across task-related brain regions. Postmenopausal women showed enhanced bilateral hippocampal connectivity relative to premenopausal and perimenopausal women. Across women, lower 17β-estradiol concentrations were related to more pronounced alterations in hippocampal connectivity and poorer performance on a subsequent memory retrieval task, strongly implicating sex steroids in the regulation of this circuitry. Finally, subgroup analyses revealed that high-performing postmenopausal women (relative to low and middle performers) exhibited a pattern of brain activity akin to premenopausal women. Together, these findings underscore the importance of considering reproductive stage, not simply chronological age, to identify neuronal and cognitive changes that unfold in the middle decades of life. In keeping with preclinical studies, these human findings suggest that the decline in ovarian estradiol production during menopause plays a significant role in shaping memory circuitry. SIGNIFICANCE STATEMENT Maintaining intact memory function with age is one of the greatest public health challenges of our time, and women have an increased risk for memory disorders relative to men later in life. We studied adults early in the aging process, as women transition into menopause, to identify neuronal and cognitive changes that unfold in the middle decades of life. Results demonstrate regional and network-level differences in memory encoding-related activity as a function of women's reproductive stage, independent of chronological age. Analyzing data without regard to sex or menopausal status obscured group differences in circuit-level neural strategies associated with successful memory retrieval. These findings suggest that early changes in memory circuitry are evident decades before the age range traditionally targeted by cognitive neuroscience of aging studies.
Collapse
|
112
|
Snyder HM, Asthana S, Bain L, Brinton R, Craft S, Dubal DB, Espeland MA, Gatz M, Mielke MM, Raber J, Rapp PR, Yaffe K, Carrillo MC. Sex biology contributions to vulnerability to Alzheimer's disease: A think tank convened by the Women's Alzheimer's Research Initiative. Alzheimers Dement 2016; 12:1186-1196. [PMID: 27692800 DOI: 10.1016/j.jalz.2016.08.004] [Citation(s) in RCA: 177] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 08/17/2016] [Indexed: 01/29/2023]
Abstract
More than 5 million Americans are living with Alzheimer's disease (AD) today, and nearly two-thirds of Americans with AD are women. This sex difference may be due to the higher longevity women generally experience; however, increasing evidence suggests that longevity alone is not a sufficient explanation and there may be other factors at play. The Alzheimer's Association convened an expert think tank to focus on the state of the science and level of evidence around gender and biological sex differences for AD, including the knowledge gaps and areas of science that need to be more fully addressed. This article summarizes the think tank discussion, moving forward a research agenda and funding program to better understand the biological underpinnings of sex- and gender-related disparities of risk for AD.
Collapse
Affiliation(s)
- Heather M Snyder
- Division of Medical & Scientific Relations, Alzheimer's Association, Chicago, IL, USA.
| | - Sanjay Asthana
- Department of Medicine, University of Wisconsin School of Medicine, Madison, WI, USA
| | - Lisa Bain
- Independent Science Writer, Philadelphia, PA, USA
| | - Roberta Brinton
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, USA; Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA; Department of Neurology, University of Southern California, Los Angeles, CA, USA
| | - Suzanne Craft
- Department of Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Dena B Dubal
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Mark A Espeland
- Department of Biostatistical Science, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Margaret Gatz
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Michelle M Mielke
- Division of Epidemiology, Department of Health Sciences Research and Neurology, Mayo Clinic, Rochester, MN, USA
| | - Jacob Raber
- Departments of Behavioral Neuroscience, Neurology, and Radiation Medicine, Oregon Health & Science University, Portland, OR, USA; Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR, USA
| | - Peter R Rapp
- Laboratory of Behavioral Neuroscience, Neurocognitive Aging Section, National Institute on Aging Intramural Research Program, Baltimore, MD, USA
| | - Kristine Yaffe
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA; Department of Psychiatry, University of California, San Francisco, San Francisco, CA, USA
| | - Maria C Carrillo
- Division of Medical & Scientific Relations, Alzheimer's Association, Chicago, IL, USA
| |
Collapse
|
113
|
Lan YL, Zou S, Zhang C, Li J, Xu Y, Li S. Update on the effect of estradiol in postmenopause women with Alzheimer's disease: a systematic review. Acta Neurol Belg 2016; 116:249-57. [PMID: 26931740 DOI: 10.1007/s13760-015-0593-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 12/25/2015] [Indexed: 02/06/2023]
Abstract
Estradiol (E2) has been used in the treatment of Alzheimer's disease (AD) for many years but with various responses. Evidence from clinical studies, randomized clinical trials (RCTs), and observational studies further underscores the importance of E2 in postmenopause women diagnosed with AD. The purpose of this article is to review all clinical trials to date focusing on the E2 in AD patients to explore the evidence regarding use of E2 in AD treatments. To achieve this objective, clinical studies regarding E2 levels in AD patients and RCTs assessing AD treatment in postmenopause women were identified through searches of MEDLINE, The Cochrane Library, EMBASE, Web of Science, Ovid, and Google Scholar. E2 has demonstrated good therapeutic effectiveness in AD patients, however, further larger scale, double-blind RCTs are required before a definitive conclusion can be reached and the results need to be compared with other drugs. This update reviews the newest clinical information regarding the role of E2 in postmenopause women with AD. To our knowledge, this is the only systematic review of this area.
Collapse
Affiliation(s)
- Yu-Long Lan
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhong Shan Road, Dalian City, 116011, People's Republic of China
- Department of Physiology, Dalian Medical University, 9 Western District, Lvshun South Road, Dalian City, 116044, People's Republic of China
| | - Shuang Zou
- Department of Physiology, Dalian Medical University, 9 Western District, Lvshun South Road, Dalian City, 116044, People's Republic of China
| | - Changfu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhong Shan Road, Dalian City, 116011, People's Republic of China
| | - Jun Li
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhong Shan Road, Dalian City, 116011, People's Republic of China
| | - Yinghui Xu
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, No. 222 Zhong Shan Road, Dalian City, 116011, People's Republic of China.
| | - Shao Li
- Department of Physiology, Dalian Medical University, 9 Western District, Lvshun South Road, Dalian City, 116044, People's Republic of China.
| |
Collapse
|
114
|
Pellegrini E, Diotel N, Vaillant-Capitaine C, Pérez Maria R, Gueguen MM, Nasri A, Cano Nicolau J, Kah O. Steroid modulation of neurogenesis: Focus on radial glial cells in zebrafish. J Steroid Biochem Mol Biol 2016; 160:27-36. [PMID: 26151741 DOI: 10.1016/j.jsbmb.2015.06.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 06/01/2015] [Accepted: 06/16/2015] [Indexed: 10/23/2022]
Abstract
Estrogens are known as steroid hormones affecting the brain in many different ways and a wealth of data now document effects on neurogenesis. Estrogens are provided by the periphery but can also be locally produced within the brain itself due to local aromatization of circulating androgens. Adult neurogenesis is described in all vertebrate species examined so far, but comparative investigations have brought to light differences between vertebrate groups. In teleost fishes, the neurogenic activity is spectacular and adult stem cells maintain their mitogenic activity in many proliferative areas within the brain. Fish are also quite unique because brain aromatase expression is limited to radial glia cells, the progenitor cells of adult fish brain. The zebrafish has emerged as an interesting vertebrate model to elucidate the cellular and molecular mechanisms of adult neurogenesis, and notably its modulation by steroids. The main objective of this review is to summarize data related to the functional link between estrogens production in the brain and neurogenesis in fish. First, we will demonstrate that the brain of zebrafish is an endogenous source of steroids and is directly targeted by local and/or peripheral steroids. Then, we will present data demonstrating the progenitor nature of radial glial cells in the brain of adult fish. Next, we will emphasize the role of estrogens in constitutive neurogenesis and its potential contribution to the regenerative neurogenesis. Finally, the negative impacts on neurogenesis of synthetic hormones used in contraceptive pills production and released in the aquatic environment will be discussed.
Collapse
Affiliation(s)
- Elisabeth Pellegrini
- Inserm U1085, Université de Rennes 1, Research Institute in Health, Environment and Occupation, 35000 Rennes, France.
| | - Nicolas Diotel
- Inserm U1085, Université de Rennes 1, Research Institute in Health, Environment and Occupation, 35000 Rennes, France; Inserm UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI), plateforme CYROI, Sainte-Clotilde F-97490, France; Université de La Réunion, UMR 1188, Sainte-Clotilde F-97490, France
| | - Colette Vaillant-Capitaine
- Inserm U1085, Université de Rennes 1, Research Institute in Health, Environment and Occupation, 35000 Rennes, France
| | - Rita Pérez Maria
- Inserm U1085, Université de Rennes 1, Research Institute in Health, Environment and Occupation, 35000 Rennes, France; Laboratorio de Ictiología, Instituto Nacional de Limnología (INALI. CONICET-UNL), Paraje El Pozo, Ciudad Universitaria UNL, 3000 Santa Fe, Argentina
| | - Marie-Madeleine Gueguen
- Inserm U1085, Université de Rennes 1, Research Institute in Health, Environment and Occupation, 35000 Rennes, France
| | - Ahmed Nasri
- Inserm U1085, Université de Rennes 1, Research Institute in Health, Environment and Occupation, 35000 Rennes, France; Laboratoire de Biosurveillance de l'Environnement, Unité d'Ecologie côtière et d'Ecotoxicologie, Faculté des Sciences de Bizerte, Zarzouna 7021, Tunisia
| | - Joel Cano Nicolau
- Inserm U1085, Université de Rennes 1, Research Institute in Health, Environment and Occupation, 35000 Rennes, France
| | - Olivier Kah
- Inserm U1085, Université de Rennes 1, Research Institute in Health, Environment and Occupation, 35000 Rennes, France
| |
Collapse
|
115
|
Menze ET, Esmat A, Tadros MG, Khalifa AE, Abdel-Naim AB. Genistein improves sensorimotor gating: Mechanisms related to its neuroprotective effects on the striatum. Neuropharmacology 2016; 105:35-46. [PMID: 26764242 DOI: 10.1016/j.neuropharm.2016.01.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 12/21/2015] [Accepted: 01/04/2016] [Indexed: 12/15/2022]
Abstract
Huntington's disease (HD) is a neurodegenerative disorder, characterized by selective atrophy in the striatum, particularly the medium spiny GABAergic efferent neurons. This results in striatal sensorimotor gating deficits. Systemic administration of 3-nitropropionic acid (3-NPA) produces selective lesions mimicking those of HD. Males were found to be more susceptible to 3-NPA-induced neurotoxicity than females, suggesting neuroprotective effects of estrogens. Phytoestrogens, including genistein, are good estrogenic alternatives that keep their beneficial effects on non-reproductive organs and lack the potential hazardous side effects. The current study was designed to investigate the potential beneficial effects of genistein in 3-NPA-induced HD in ovariectomized rats. Results showed that 3-NPA (20 mg/kg) administration caused significant disruption of the rats' locomotor activity and prepulse inhibition. In addition, it decreased striatal ATP levels and increased oxidative stress, inflammatory and apoptotic markers with striatal focal hemorrhage and gliosis. Pretreatment with 17β-estradiol (2.5 mg/kg) or genistein (20 mg/kg) led to a significant improvement of behavioral parameters, increased ATP production, decreased oxidative stress, attenuated inflammation and apoptosis. Therefore, this study suggests potential neuroprotective effects of genistein in ovariectomized rats challenged with 3-NPA.
Collapse
Affiliation(s)
- Esther T Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ahmed Esmat
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mariane G Tadros
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Amani E Khalifa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ashraf B Abdel-Naim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
116
|
Genzel L, Bäurle A, Potyka A, Wehrle R, Adamczyk M, Friess E, Steiger A, Dresler M. Diminished nap effects on memory consolidation are seen under oral contraceptive use. Neuropsychobiology 2016; 70:253-261. [PMID: 25720656 DOI: 10.1159/000369022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 10/10/2014] [Indexed: 11/19/2022]
Abstract
Many young females take exogenous hormones as oral contraceptive (OC), a condition rarely controlled for in studies on sleep and memory consolidation even though sex hormones influence consolidation. This study investigated the effects of OCs on sleep-related consolidation of a motor and declarative task, utilizing a daytime nap protocol. Fifteen healthy, young females taking OCs came to the sleep lab for three different conditions: nap with previous learning, wake with previous learning and nap without learning. They underwent each condition twice, once during the "pill-active" weeks and once during the "pill-free" week, resulting in 6 visits. In all conditions, participants showed a significant off-line consolidation effect, independent of pill week or nap/wake condition. There were no significant differences in sleep stage duration, spindle activity or spectral EEG frequency bands between naps with or without the learning condition. The present data showed a significant off-line enhancement in memory irrespective of potential beneficial effects of a nap. In comparison to previous studies, this may suggest that the use of OCs may enhance off-line memory consolidation in motor and verbal tasks per se. These results stress the importance to control for the use of OCs in studies focusing on memory performance.
Collapse
|
117
|
Zhao L, Mao Z, Woody SK, Brinton RD. Sex differences in metabolic aging of the brain: insights into female susceptibility to Alzheimer's disease. Neurobiol Aging 2016; 42:69-79. [PMID: 27143423 DOI: 10.1016/j.neurobiolaging.2016.02.011] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 02/08/2016] [Accepted: 02/10/2016] [Indexed: 10/22/2022]
Abstract
Despite recent advances in the understanding of clinical aspects of sex differences in Alzheimer's disease (AD), the underlying mechanisms, for instance, how sex modifies AD risk and why the female brain is more susceptible to AD, are not clear. The purpose of this study is to elucidate sex disparities in brain aging profiles focusing on 2 major areas-energy and amyloid metabolism-that are most significantly affected in preclinical development of AD. Total RNA isolated from hippocampal tissues of both female and male 129/C57BL/6 mice at ages of 6, 9, 12, or 15 months were comparatively analyzed by custom-designed Taqman low-density arrays for quantitative real-time polymerase chain reaction detection of a total of 182 genes involved in a broad spectrum of biological processes modulating energy production and amyloid homeostasis. Gene expression profiles revealed substantial differences in the trajectory of aging changes between female and male brains. In female brains, 44.2% of genes were significantly changed from 6 months to 9 months and two-thirds showed downregulation. In contrast, in male brains, only 5.4% of genes were significantly altered at this age transition. Subsequent changes in female brains were at a much smaller magnitude, including 10.9% from 9 months to 12 months and 6.1% from 12 months to 15 months. In male brains, most changes occurred from 12 months to 15 months and the majority were upregulated. Furthermore, gene network analysis revealed that clusterin appeared to serve as a link between the overall decreased bioenergetic metabolism and increased amyloid dyshomeostasis associated with the earliest transition in female brains. Together, results from this study indicate that: (1) female and male brains follow profoundly dissimilar trajectories as they age; (2) female brains undergo age-related changes much earlier than male brains; (3) early changes in female brains signal the onset of a hypometabolic phenotype at risk for AD. These findings provide a mechanistic rationale for female susceptibility to AD and suggest a potential window of opportunity for AD prevention and risk reduction in women.
Collapse
Affiliation(s)
- Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA; Neuroscience Graduate Program, University of Kansas, Lawrence, KS, USA; Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA.
| | - Zisu Mao
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Sarah K Woody
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Roberta D Brinton
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
118
|
Mapping of brain lipid binding protein (Blbp) in the brain of adult zebrafish, co-expression with aromatase B and links with proliferation. Gene Expr Patterns 2016; 20:42-54. [DOI: 10.1016/j.gep.2015.11.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 09/25/2015] [Accepted: 11/10/2015] [Indexed: 01/05/2023]
|
119
|
Souza CS, Paulsen BS, Devalle S, Lima Costa S, Borges HL, Rehen SK. Commitment of human pluripotent stem cells to a neural lineage is induced by the pro-estrogenic flavonoid apigenin. ACTA ACUST UNITED AC 2015. [DOI: 10.3402/arb.v2.29244] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
120
|
Espeland MA, Brinton RD, Hugenschmidt C, Manson JE, Craft S, Yaffe K, Weitlauf J, Vaughan L, Johnson KC, Padula CB, Jackson RD, Resnick SM. Impact of Type 2 Diabetes and Postmenopausal Hormone Therapy on Incidence of Cognitive Impairment in Older Women. Diabetes Care 2015; 38:2316-24. [PMID: 26486190 PMCID: PMC4657616 DOI: 10.2337/dc15-1385] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/17/2015] [Indexed: 02/03/2023]
Abstract
OBJECTIVE In older women, higher levels of estrogen may exacerbate the increased risk for cognitive impairment conveyed by diabetes. We examined whether the effect of postmenopausal hormone therapy (HT) on cognitive impairment incidence differs depending on type 2 diabetes. RESEARCH DESIGN AND METHODS The Women's Health Initiative (WHI) randomized clinical trials assigned women to HT (0.625 mg/day conjugated equine estrogens with or without [i.e., unopposed] 2.5 mg/day medroxyprogesterone acetate) or matching placebo for an average of 4.7-5.9 years. A total of 7,233 women, aged 65-80 years, were classified according to type 2 diabetes status and followed for probable dementia and cognitive impairment (mild cognitive impairment or dementia). RESULTS Through a maximum of 18 years of follow-up, women with diabetes had increased risk of probable dementia (hazard ratio [HR] 1.54 [95% CI 1.16-2.06]) and cognitive impairment (HR 1.83 [1.50-2.23]). The combination of diabetes and random assignment to HT increased their risk of dementia (HR 2.12 [1.47-3.06]) and cognitive impairment (HR 2.20 [1.70-2.87]) compared with women without these conditions, interaction P = 0.09 and P = 0.08. These interactions appeared to be limited to women assigned to unopposed conjugated equine estrogens. CONCLUSIONS These analyses provide additional support to a prior report that higher levels of estrogen may exacerbate risks that type 2 diabetes poses for cognitive function in older women. The role estrogen plays in suppressing non-glucose-based energy sources in the brain may explain this interaction.
Collapse
Affiliation(s)
- Mark A Espeland
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC
| | - Roberta Diaz Brinton
- Departments of Pharmacology and Pharmaceutical Sciences, Biomedical Engineering, and Neurology, University of Southern California, Los Angeles, CA
| | | | - JoAnn E Manson
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Suzanne Craft
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Kristine Yaffe
- Departments of Epidemiology and Biostatistics, Psychiatry, and Neurology, University of California, San Francisco, San Francisco, CA
| | - Julie Weitlauf
- Veterans Affairs Palo Alto Health Care System, Stanford University School of Medicine, Stanford, CA Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA
| | - Leslie Vaughan
- Department of Social Sciences and Health Policy, Wake Forest School of Medicine, Winston-Salem, NC
| | - Karen C Johnson
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Claudia B Padula
- Veterans Affairs Palo Alto Health Care System, Stanford University School of Medicine, Stanford, CA Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA
| | - Rebecca D Jackson
- Department of Internal Medicine, Ohio State University, Columbus, OH
| | - Susan M Resnick
- Laboratory of Behavioral Neuroscience, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD
| |
Collapse
|
121
|
Simone J, Bogue EA, Bhatti DL, Day LE, Farr NA, Grossman AM, Holmes PV. Ethinyl estradiol and levonorgestrel alter cognition and anxiety in rats concurrent with a decrease in tyrosine hydroxylase expression in the locus coeruleus and brain-derived neurotrophic factor expression in the hippocampus. Psychoneuroendocrinology 2015; 62:265-78. [PMID: 26352480 DOI: 10.1016/j.psyneuen.2015.08.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 08/18/2015] [Accepted: 08/18/2015] [Indexed: 12/31/2022]
Abstract
In the United States, more than ten million women use contraceptive hormones. Ethinyl estradiol and levonorgestrel have been mainstay contraceptive hormones for the last four decades. Surprisingly, there is scant information regarding their action on the central nervous system and behavior. Intact female rats received three weeks of subcutaneous ethinyl estradiol (10 or 30μg/rat/day), levonorgestrel (20 or 60μg/rat/day), a combination of both (10/20μg/rat/day and 30/60μg/rat/day), or vehicle. Subsequently, the rats were tested in three versions of the novel object recognition test to assess learning and memory, and a battery of tests for anxiety-like behavior. Serum estradiol and ovarian weights were measured. All treatment groups exhibited low endogenous 17β-estradiol levels at the time of testing. Dose-dependent effects of drug treatment manifested in both cognitive and anxiety tests. All low dose drugs decreased anxiety-like behavior and impaired performance on novel object recognition. In contrast, the high dose ethinyl estradiol increased anxiety-like behavior and improved performance in cognitive testing. In the cell molecular analyses, low doses of all drugs induced a decrease in tyrosine hydroxylase mRNA and protein in the locus coeruleus. At the same time, low doses of ethinyl estradiol and ethinyl estradiol/levonorgestrel increased galanin protein in this structure. Consistent with the findings above, the low dose treatments of ethinyl estradiol and combination ethinyl estradiol/levonorgestrel reduced brain-derived neurotrophic factor mRNA in the hippocampus. These effects of ethinyl estradiol 10μg alone and in combination with levonorgestrel 20μg suggest a diminution of norepinephrine input into the hippocampus resulting in a decline in learning and memory.
Collapse
Affiliation(s)
- Jean Simone
- Neuroscience, Biomedical and Health Sciences Institute, University of Georgia, 150 Paul D. Coverdell Center, Athens, GA 30602, USA.
| | - Elizabeth A Bogue
- Neuroscience, Biomedical and Health Sciences Institute, University of Georgia, 150 Paul D. Coverdell Center, Athens, GA 30602, USA.
| | - Dionnet L Bhatti
- Neuroscience, Biomedical and Health Sciences Institute, University of Georgia, 150 Paul D. Coverdell Center, Athens, GA 30602, USA.
| | - Laura E Day
- Neuroscience, Biomedical and Health Sciences Institute, University of Georgia, 150 Paul D. Coverdell Center, Athens, GA 30602, USA.
| | - Nathan A Farr
- Neuroscience, Biomedical and Health Sciences Institute, University of Georgia, 150 Paul D. Coverdell Center, Athens, GA 30602, USA.
| | - Anna M Grossman
- Neuroscience, Biomedical and Health Sciences Institute, University of Georgia, 150 Paul D. Coverdell Center, Athens, GA 30602, USA.
| | - Philip V Holmes
- Neuroscience, Biomedical and Health Sciences Institute, University of Georgia, 150 Paul D. Coverdell Center, Athens, GA 30602, USA; Psychology, University of Georgia, 125 Baldwin Street, Athens, GA 30602, USA.
| |
Collapse
|
122
|
Corvino V, Di Maria V, Marchese E, Lattanzi W, Biamonte F, Michetti F, Geloso MC. Estrogen administration modulates hippocampal GABAergic subpopulations in the hippocampus of trimethyltin-treated rats. Front Cell Neurosci 2015; 9:433. [PMID: 26594149 PMCID: PMC4633568 DOI: 10.3389/fncel.2015.00433] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/15/2015] [Indexed: 12/13/2022] Open
Abstract
Given the well-documented involvement of estrogens in the modulation of hippocampal functions in both physiological and pathological conditions, the present study investigates the effects of 17-beta estradiol (E2) administration in the rat model of hippocampal neurodegeneration induced by trimethyltin (TMT) administration (8 mg/kg), characterized by loss of pyramidal neurons in CA1, CA3/hilus hippocampal subfields, associated with astroglial and microglial activation, seizures and cognitive impairment. After TMT/saline treatment, ovariectomized animals received two doses of E2 (0.2 mg/kg intra-peritoneal) or vehicle, and were sacrificed 48 h or 7 days after TMT-treatment. Our results indicate that in TMT-treated animals E2 administration induces the early (48 h) upregulation of genes involved in neuroprotection and synaptogenesis, namely Bcl2, trkB, cadherin 2 and cyclin-dependent-kinase-5. Increased expression levels of glutamic acid decarboxylase (gad) 67, neuropeptide Y (Npy), parvalbumin, Pgc-1α and Sirtuin 1 genes, the latter involved in parvalbumin (PV) synthesis, were also evident. Unbiased stereology performed on rats sacrificed 7 days after TMT treatment showed that although E2 does not significantly influence the extent of TMT-induced neuronal death, significantly enhances the TMT-induced modulation of GABAergic interneuron population size in selected hippocampal subfields. In particular, E2 administration causes, in TMT-treated rats, a significant increase in the number of GAD67-expressing interneurons in CA1 stratum oriens, CA3 pyramidal layer, hilus and dentate gyrus, accompanied by a parallel increase in NPY-expressing cells, essentially in the same regions, and of PV-positive cells in CA1 pyramidal layer. The present results add information concerning the role of in vivo E2 administration on mechanisms involved in cellular plasticity in the adult brain.
Collapse
Affiliation(s)
- Valentina Corvino
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| | - Valentina Di Maria
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| | - Elisa Marchese
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| | - Wanda Lattanzi
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| | - Filippo Biamonte
- Institute of Histology and Embryology, Università Cattolica del Sacro Cuore Rome, Italy
| | - Fabrizio Michetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| | - Maria Concetta Geloso
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| |
Collapse
|
123
|
Zhao L, Woody SK, Chhibber A. Estrogen receptor β in Alzheimer's disease: From mechanisms to therapeutics. Ageing Res Rev 2015; 24:178-90. [PMID: 26307455 DOI: 10.1016/j.arr.2015.08.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 08/04/2015] [Accepted: 08/17/2015] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) disproportionally affects women and men. The female susceptibility for AD has been largely associated with the loss of ovarian sex hormones during menopause. This review examines the current understanding of the role of estrogen receptor β (ERβ) in the regulation of neurological health and its implication in the development and intervention of AD. Since its discovery in 1996, research conducted over the last 15-20 years has documented a great deal of evidence indicating that ERβ plays a pivotal role in a broad spectrum of brain activities from development to aging. ERβ genetic polymorphisms have been associated with cognitive impairment and increased risk for AD predominantly in women. The role of ERβ in the intervention of AD has been demonstrated by the alteration of AD pathology in response to treatment with ERβ-selective modulators in transgenic models that display pronounced plaque and tangle histopathological presentations as well as learning and memory deficits. Future studies that explore the potential interactions between ERβ signaling and the genetic isoforms of human apolipoprotein E (APOE) in brain aging and development of AD-risk phenotype are critically needed. The current trend of lost-in-translation in AD drug development that has primarily been based on early-onset familial AD (FAD) models underscores the urgent need for novel models that recapitulate the etiology of late-onset sporadic AD (SAD), the most common form of AD representing more than 95% of the current human AD population. Combining the use of FAD-related models that generally have excellent face validity with SAD-related models that hold more reliable construct validity would together increase the predictive validity of preclinical findings for successful translation into humans.
Collapse
Affiliation(s)
- Liqin Zhao
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA; Neuroscience Graduate Program, University of Kansas, Lawrence, KS 66045, USA.
| | - Sarah K Woody
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| | - Anindit Chhibber
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, Lawrence, KS 66045, USA
| |
Collapse
|
124
|
Wroolie TE, Kenna HA, Williams KE, Rasgon NL. Cognitive Effects of Hormone Therapy Continuation or Discontinuation in a Sample of Women at Risk for Alzheimer Disease. Am J Geriatr Psychiatry 2015; 23. [PMID: 26209223 PMCID: PMC4654994 DOI: 10.1016/j.jagp.2015.05.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Use of estrogen-based hormone therapy (HT) as a protection from cognitive decline and Alzheimer disease (AD) is controversial, although cumulative data support HT use when initiated close to menopause onset with estrogen formulations containing 17β-estradiol preferable to conjugated equine estrogen formulations. Little is known regarding specific populations of women who may derive benefit from HT. METHODS Women with heightened risk for AD (aged 49-69), all of whom were taking HT for at least 1 year and most of whom initiated HT close to menopause onset, underwent cognitive assessment followed by randomization to continue or discontinue HT. Assessments were repeated at 2 years after randomization. RESULTS Women who continued HT performed better on cognitive domains composed of measures of verbal memory and combined attention, working memory, and processing speed measures. Women who used 17β-estradiol versus conjugated equine estrogen, whether randomized to continue or discontinue HT, showed better verbal memory performance at the 2-year follow-up assessment. An interaction was also found with HT randomization and family history of AD in a first-degree relative. All female offspring of patients with AD declined in verbal memory; however, women who continued HT declined less than women who discontinued HT. Women without a first-degree relative with AD showed verbal memory improvement (likely because of practice effects) with continuance and declined with discontinuance of HT. CONCLUSION Continuation of HT use appears to protect cognition in women with heightened risk for AD when initiated close to menopause onset.
Collapse
Affiliation(s)
- Tonita E. Wroolie
- Stanford Center for Neuroscience in Women's Health, Department of Psychiatry and Behavioral Sciences, 401 Quarry Road, Stanford University School of Medicine, Stanford, CA 94303-5723, USA
| | - Heather A. Kenna
- Stanford Center for Neuroscience in Women's Health, Department of Psychiatry and Behavioral Sciences, 401 Quarry Road, Stanford University School of Medicine, Stanford, CA 94303-5723, USA
| | - Katherine E. Williams
- Stanford Center for Neuroscience in Women's Health, Department of Psychiatry and Behavioral Sciences, 401 Quarry Road, Stanford University School of Medicine, Stanford, CA 94303-5723, USA
| | - Natalie L. Rasgon
- Stanford Center for Neuroscience in Women's Health, Department of Psychiatry and Behavioral Sciences, 401 Quarry Road, Stanford University School of Medicine, Stanford, CA 94303-5723, USA
| |
Collapse
|
125
|
Xu Y, Liu Z, Song X, Zhang K, Li X, Li J, Yan X, Li Y, Xie Z, Zhang H. Cerebralcare Granule® attenuates cognitive impairment in rats continuously overexpressing microRNA-30e. Mol Med Rep 2015; 12:8032-40. [PMID: 26498486 PMCID: PMC4758320 DOI: 10.3892/mmr.2015.4469] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 09/09/2015] [Indexed: 11/23/2022] Open
Abstract
Previous studies have demonstrated that dysregulation of micro (mi)RNAs is associated with the etiology of various neuropsychiatric disorders, including depression and schizophrenia. Cerebralcare Granule® (CG) is a Chinese herbal medicine, which has been reported to have an ameliorative effect on brain injury by attenuating blood-brain barrier disruption and improving hippocampal neural function. The present study aimed to evaluate the cognitive behavior of rats continuously overexpressing miRNA-30e (lenti-miRNA-30e), prior to and following the administration of CG. In addition, the mechanisms underlying the ameliorative effects of CG were investigated. The cognitive ability of the rats was assessed using an open-field test and a Morris water maze spatial reference/working memory test. A terminal deoxynucleotidyl transferase dUTP nick end labeling assay was used to detect neuronal apoptosis in the dentate gyrus of the hippocampus. Immunohistochemical analysis and western blotting were conducted to detect the expression levels of B-cell lymphoma 2 (BCL-2) and ubiquitin-conjugating enzyme 9 (UBC9), in order to examine neuronal apoptosis. The lenti-miRNA-30e rats exhibited increased signs of anxiety, depression, hyperactivity and schizophrenia, which resulted in a severe impairment in cognitive ability. Furthermore, in the dentate gyrus of these rats, the expression levels of BCL-2 and UBC9 were reduced and apoptosis was increased. The administration of CG alleviated cognitive impairment, enhanced the expression levels of BCL-2 and UBC9, and reduced apoptosis in the dentate gyrus in the lenti-miRNA-30e rats. No significant differences were detected in behavioral indicators between the lenti-miRNA-30e rats treated with CG and the normal controls. These findings suggested that CG exerts a potent therapeutic effect, conferred by its ability to enhance the expression levels of BCL-2 and UBC9, which inhibits the apoptotic process in neuronal cells. Therefore, CG may be considered a potential therapeutic strategy for the treatment of cognitive impairment in mental disorders.
Collapse
Affiliation(s)
- Yong Xu
- Department of Psychiatry, First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Zhifen Liu
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xi Song
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Kerang Zhang
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xingrong Li
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jianhong Li
- Department of Psychiatry, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Xu Yan
- Department of Psychiatry, First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Yuan Li
- Department of Interventional Radiography, Heping Hospital of Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Zhongchen Xie
- Laboratory Animal Facility Biomedical Analysis Center, Tsinghua University, Beijing 100084, P.R. China
| | - Hui Zhang
- Department of Radiology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
126
|
Mathews SB, Epperson CN. Neuropsychiatric Disorders Among Aging Women: Assessing Risk Factors and Tailoring Treatment. Curr Behav Neurosci Rep 2015. [DOI: 10.1007/s40473-015-0057-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
127
|
Kindler J, Weickert CS, Skilleter AJ, Catts SV, Lenroot R, Weickert TW. Selective Estrogen Receptor Modulation Increases Hippocampal Activity during Probabilistic Association Learning in Schizophrenia. Neuropsychopharmacology 2015; 40:2388-97. [PMID: 25829142 PMCID: PMC4538353 DOI: 10.1038/npp.2015.88] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 02/14/2015] [Accepted: 03/09/2015] [Indexed: 02/06/2023]
Abstract
People with schizophrenia show probabilistic association learning impairment in conjunction with abnormal neural activity. The selective estrogen receptor modulator (SERM) raloxifene preserves neural activity during memory in healthy older men and improves memory in schizophrenia. Here, we tested the extent to which raloxifene modifies neural activity during learning in schizophrenia. Nineteen people with schizophrenia participated in a twelve-week randomized, double-blind, placebo-controlled, cross-over adjunctive treatment trial of the SERM raloxifene administered orally at 120 mg daily to assess brain activity during probabilistic association learning using functional magnetic resonance imaging (fMRI). Raloxifene improved probabilistic association learning and significantly increased fMRI BOLD activity in the hippocampus and parahippocampal gyrus relative to placebo. A separate region of interest confirmatory analysis in 21 patients vs 36 healthy controls showed a positive association between parahippocampal neural activity and learning in patients, but no such relationship in the parahippocampal gyrus of healthy controls. Thus, selective estrogen receptor modulation by raloxifene concurrently increases activity in the parahippocampal gyrus and improves probabilistic association learning in schizophrenia. These results support a role for estrogen receptor modulation of mesial temporal lobe neural activity in the remediation of learning disabilities in both men and women with schizophrenia.
Collapse
Affiliation(s)
- Jochen Kindler
- School of Psychiatry, University of New South Wales, Randwick, NSW, Australia,Neuroscience Research Australia, Randwick, NSW, Australia,Department of Psychiatric Neurophysiology, University of Bern, Bern, Switzerland
| | - Cynthia Shannon Weickert
- School of Psychiatry, University of New South Wales, Randwick, NSW, Australia,Neuroscience Research Australia, Randwick, NSW, Australia,Schizophrenia Research Institute, Darlinghurst, NSW, Australia
| | - Ashley J Skilleter
- School of Psychiatry, University of New South Wales, Randwick, NSW, Australia,Neuroscience Research Australia, Randwick, NSW, Australia,Schizophrenia Research Institute, Darlinghurst, NSW, Australia
| | - Stanley V Catts
- School of Medical Science, University of Queensland, Brisbane, QLD, Australia
| | - Rhoshel Lenroot
- School of Psychiatry, University of New South Wales, Randwick, NSW, Australia,Neuroscience Research Australia, Randwick, NSW, Australia,Schizophrenia Research Institute, Darlinghurst, NSW, Australia
| | - Thomas W Weickert
- School of Psychiatry, University of New South Wales, Randwick, NSW, Australia,Neuroscience Research Australia, Randwick, NSW, Australia,Schizophrenia Research Institute, Darlinghurst, NSW, Australia,School of Psychiatry, University of New South Wales, Neuroscience Research Australia, Barker Street, Randwick, NSW 2031, Australia, Tel: +61 2 9399 1730, Fax: +61 2 9399 1034, E-mail:
| |
Collapse
|
128
|
Shum C, Macedo SC, Warre-Cornish K, Cocks G, Price J, Srivastava DP. Utilizing induced pluripotent stem cells (iPSCs) to understand the actions of estrogens in human neurons. Horm Behav 2015; 74:228-42. [PMID: 26143621 PMCID: PMC4579404 DOI: 10.1016/j.yhbeh.2015.06.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 06/11/2015] [Accepted: 06/25/2015] [Indexed: 01/05/2023]
Abstract
This article is part of a Special Issue "Estradiol and Cognition". Over recent years tremendous progress has been made towards understanding the molecular and cellular mechanism by which estrogens exert enhancing effects on cognition, and how they act as a neuroprotective or neurotrophic agent in disease. Currently, much of this work has been carried out in animal models with only a limited number of studies using native human tissue or cells. Recent advances in stem cell technology now make it possible to reprogram somatic cells from humans into induced pluripotent stem cells (iPSCs), which can subsequently be differentiated into neurons of specific lineages. Importantly, the reprogramming of cells allows for the generation of iPSCs that retain the genetic "makeup" of the donor. Therefore, it is possible to generate iPSC-derived neurons from patients diagnosed with specific diseases, that harbor the complex genetic background associated with the disorder. Here, we review the iPSC technology and how it's currently being used to model neural development and neurological diseases. Furthermore, we explore whether this cellular system could be used to understand the role of estrogens in human neurons, and present preliminary data in support of this. We further suggest that the use of iPSC technology offers a novel system to not only further understand estrogens' effects in human cells, but also to investigate the mechanism by which estrogens are beneficial in disease. Developing a greater understanding of these mechanisms in native human cells will also aid in the development of safer and more effective estrogen-based therapeutics.
Collapse
Affiliation(s)
- Carole Shum
- Department of Basic and Clinical Neuroscience, Cell and Behaviour Unit, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Sara C Macedo
- Department of Basic and Clinical Neuroscience, Cell and Behaviour Unit, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK; Faculty of Engineering, Universidade do Porto, 4200-465 Porto, Portugal
| | - Katherine Warre-Cornish
- Department of Basic and Clinical Neuroscience, Cell and Behaviour Unit, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Graham Cocks
- Department of Basic and Clinical Neuroscience, Cell and Behaviour Unit, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Jack Price
- Department of Basic and Clinical Neuroscience, Cell and Behaviour Unit, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK
| | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, Cell and Behaviour Unit, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 8AF, UK.
| |
Collapse
|
129
|
Abstract
Perimenopause is a midlife transition state experienced by women that occurs in the context of a fully functioning neurological system and results in reproductive senescence. Although primarily viewed as a reproductive transition, the symptoms of perimenopause are largely neurological in nature. Neurological symptoms that emerge during perimenopause are indicative of disruption in multiple estrogen-regulated systems (including thermoregulation, sleep, circadian rhythms and sensory processing) and affect multiple domains of cognitive function. Estrogen is a master regulator that functions through a network of estrogen receptors to ensure that the brain effectively responds at rapid, intermediate and long timescales to regulate energy metabolism in the brain via coordinated signalling and transcriptional pathways. The estrogen receptor network becomes uncoupled from the bioenergetic system during the perimenopausal transition and, as a corollary, a hypometabolic state associated with neurological dysfunction can develop. For some women, this hypometabolic state might increase the risk of developing neurodegenerative diseases later in life. The perimenopausal transition might also represent a window of opportunity to prevent age-related neurological diseases. This Review considers the importance of neurological symptoms in perimenopause in the context of their relationship to the network of estrogen receptors that control metabolism in the brain.
Collapse
Affiliation(s)
- Roberta D Brinton
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA
| | - Jia Yao
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA
| | - Fei Yin
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA
| | - Wendy J Mack
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, 1975 Zonal Avenue, Los Angeles, CA 90089, USA
| | - Enrique Cadenas
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, USA
| |
Collapse
|
130
|
|
131
|
Mannella P, Simoncini T, Genazzani AR. Estrogens and progestins: molecular effects on brain cells. Horm Mol Biol Clin Investig 2015; 4:609-13. [PMID: 25961237 DOI: 10.1515/hmbci.2010.078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Accepted: 12/06/2010] [Indexed: 11/15/2022]
Abstract
Sex steroids are known to regulate brain function and their role is so important that several diseases are strictly correlated with the onset of menopause when estrogen-progesterone deficiency makes neural cells much more vulnerable to toxic stimuli. Although in the past years several scientists have focused their studies on in vitro and in vivo effects of sex steroids on the brain, we are still far from complete knowledge. Indeed, contrasting results from large clinical trials have made the entire issue much more complicated. Currently we know that protective effects exerted by sex steroids depend on several factors among which the dose, the health of the cells and the type of molecule being used. In this review, we present an overview of the direct and indirect effects of estrogen and progesterone on the brain with specific focus on the molecular mechanisms by which these molecules act on neural cells.
Collapse
|
132
|
Hajali V, Sheibani V, Mahani SE, Hajializadeh Z, Shabani M, Aliabadi HP, Saadati H, Esmaeilpour K. Ovariectomy does not exacerbate the negative effects of sleep deprivation on synaptic plasticity in rats. Physiol Behav 2015; 144:73-81. [DOI: 10.1016/j.physbeh.2015.03.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 03/02/2015] [Accepted: 03/04/2015] [Indexed: 10/23/2022]
|
133
|
Yin F, Yao J, Sancheti H, Feng T, Melcangi RC, Morgan TE, Finch CE, Pike CJ, Mack WJ, Cadenas E, Brinton RD. The perimenopausal aging transition in the female rat brain: decline in bioenergetic systems and synaptic plasticity. Neurobiol Aging 2015; 36:2282-2295. [PMID: 25921624 DOI: 10.1016/j.neurobiolaging.2015.03.013] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/20/2015] [Accepted: 03/25/2015] [Indexed: 01/03/2023]
Abstract
The perimenopause is an aging transition unique to the female that leads to reproductive senescence which can be characterized by multiple neurological symptoms. To better understand potential underlying mechanisms of neurological symptoms of perimenopause, the present study determined genomic, biochemical, brain metabolic, and electrophysiological transformations that occur during this transition using a rat model recapitulating fundamental characteristics of the human perimenopause. Gene expression analyses indicated two distinct aging programs: chronological and endocrine. A critical period emerged during the endocrine transition from regular to irregular cycling characterized by decline in bioenergetic gene expression, confirmed by deficits in fluorodeoxyglucose-positron emission tomography (FDG-PET) brain metabolism, mitochondrial function, and long-term potentiation. Bioinformatic analysis predicted insulin/insulin-like growth factor 1 and adenosine monophosphate-activated protein kinase/peroxisome proliferator-activated receptor gamma coactivator 1 alpha (AMPK/PGC1α) signaling pathways as upstream regulators. Onset of acyclicity was accompanied by a rise in genes required for fatty acid metabolism, inflammation, and mitochondrial function. Subsequent chronological aging resulted in decline of genes required for mitochondrial function and β-amyloid degradation. Emergence of glucose hypometabolism and impaired synaptic function in brain provide plausible mechanisms of neurological symptoms of perimenopause and may be predictive of later-life vulnerability to hypometabolic conditions such as Alzheimer's.
Collapse
Affiliation(s)
- Fei Yin
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Jia Yao
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Harsh Sancheti
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Tao Feng
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Roberto C Melcangi
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Todd E Morgan
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Caleb E Finch
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Christian J Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Wendy J Mack
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Enrique Cadenas
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Roberta D Brinton
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
134
|
Castonguay N, Lussier M, Bugaiska A, Lord C, Bherer L. Executive functions in men and postmenopausal women. J Clin Exp Neuropsychol 2015; 37:193-208. [PMID: 25695230 DOI: 10.1080/13803395.2014.1000267] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
INTRODUCTION This study was designed to assess sex differences in older adults (55-65 years old) in executive functions and to examine the influence of hormone therapy (HT) in postmenopausal women. METHOD We have assessed task performance in memory, visuospatial, and executive functions in 29 women using HT, 29 women who never used HT, and 30 men. RESULTS Men outperformed never users in task switching and updating. HT users outperformed never users in updating. HT users outperformed never users and men in visual divided attention. DISCUSSION The present study support previous findings that sex and HT impact cognition and bring new insights on sex and HT-related differences in executive functions.
Collapse
Affiliation(s)
- Nathalie Castonguay
- a Department of Psychology , Université du Québec à Montréal , Montreal , QC , Canada
| | | | | | | | | |
Collapse
|
135
|
Menze ET, Esmat A, Tadros MG, Abdel-Naim AB, Khalifa AE. Genistein improves 3-NPA-induced memory impairment in ovariectomized rats: impact of its antioxidant, anti-inflammatory and acetylcholinesterase modulatory properties. PLoS One 2015; 10:e0117223. [PMID: 25675218 PMCID: PMC4326416 DOI: 10.1371/journal.pone.0117223] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 12/20/2014] [Indexed: 12/21/2022] Open
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder. The pre-motor symptomatic stages of the disease are commonly characterized by cognitive problems including memory loss. 3-Nitropropionic acid (3-NPA) is a mitochondrial toxin that produces selective lesions in the brain similar to that of HD and was proven to cause memory impairment in rodents. Phytoestrogens have well-established neuroprotective and memory enhancing effects with fewer side effects in comparison to estrogens. This study investigated the potential neuroprotective and memory enhancing effect of genistein (5, 10 and 20 mg/kg), a phytoestrogen, in ovariectomized rats challenged with 3-NPA (20 mg/kg). These potential effects were compared to those of 17β-estradiol (2.5 mg/kg). Systemic administration of 3-NPA for 4 consecutive days impaired locomotor activity, decreased retention latencies in the passive avoidance task, decreased striatal, cortical and hippocampal ATP levels, increased oxidative stress, acetylcholinesterase (AChE) activity, cycloxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) expressions. Pretreatment with genistein and 17β-estradiol attenuated locomotor hypoactivity, increased retention latencies in the passive avoidance task, increased ATP levels, improved the oxidative stress profile, attenuated the increase in AChE activity and decreased the expression of COX-2 and iNOS. Overall, the higher genistein dose (20 mg/kg) was the most effective. In conclusion, this study suggests neuroprotective and memory enhancing effects for genistein in a rat model of HD. These effects might be attributed to its antioxidant, anti-inflammatory and cholinesterase inhibitory activities.
Collapse
Affiliation(s)
- Esther T. Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ahmed Esmat
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mariane G. Tadros
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ashraf B. Abdel-Naim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Amani E. Khalifa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
136
|
Xu X, Gu T, Shen Q. Different effects of bisphenol-A on memory behavior and synaptic modification in intact and estrogen-deprived female mice. J Neurochem 2015; 132:572-82. [DOI: 10.1111/jnc.12998] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 11/10/2014] [Accepted: 11/13/2014] [Indexed: 11/28/2022]
Affiliation(s)
- Xiaohong Xu
- Chemistry and Life Sciences College; Zhejiang Normal University; Jinhua China
| | - Ting Gu
- Chemistry and Life Sciences College; Zhejiang Normal University; Jinhua China
| | - Qiaoqiao Shen
- Chemistry and Life Sciences College; Zhejiang Normal University; Jinhua China
| |
Collapse
|
137
|
Sellers K, Raval P, Srivastava DP. Molecular signature of rapid estrogen regulation of synaptic connectivity and cognition. Front Neuroendocrinol 2015; 36:72-89. [PMID: 25159586 DOI: 10.1016/j.yfrne.2014.08.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 08/11/2014] [Accepted: 08/14/2014] [Indexed: 12/14/2022]
Abstract
There is now a growing appreciation that estrogens are capable of rapidly activating a number of signaling cascades within the central nervous system. In addition, there are an increasing number of studies reporting that 17β-estradiol, the major biologically active estrogen, can modulate cognition within a rapid time frame. Here we review recent studies that have begun to uncover the molecular and cellular framework which contributes to estrogens ability to rapidly modulate cognition. We first describe the mechanisms by which estrogen receptors (ERs) can couple to intracellular signaling cascades, either directly, or via the transactivation of other receptors. Subsequently, we review the evidence that estrogen can rapidly modulate both neuronal function and structure in the hippocampus and the cortex. Finally, we will discuss how estrogens may influence cognitive function through the modulation of neuronal structure, and the implications this may have on the treatment of a range of brain disorders.
Collapse
Affiliation(s)
- Katherine Sellers
- Department of Basic and Clinical Neuroscience, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, UK
| | - Pooja Raval
- Department of Basic and Clinical Neuroscience, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, UK
| | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, UK.
| |
Collapse
|
138
|
Wu YWC, Du X, van den Buuse M, Hill RA. Analyzing the influence of BDNF heterozygosity on spatial memory response to 17β-estradiol. Transl Psychiatry 2015; 5:e498. [PMID: 25603414 PMCID: PMC4312832 DOI: 10.1038/tp.2014.143] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 12/02/2014] [Indexed: 12/25/2022] Open
Abstract
The recent use of estrogen-based therapies as adjunctive treatments for the cognitive impairments of schizophrenia has produced promising results; however the mechanism behind estrogen-based cognitive enhancement is relatively unknown. Brain-derived neurotrophic factor (BDNF) regulates learning and memory and its expression is highly responsive to estradiol. We recently found that estradiol modulates the expression of hippocampal parvalbumin-positive GABAergic interneurons, known to regulate neuronal synchrony and cognitive function. What is unknown is whether disruptions to the aforementioned estradiol-parvalbumin pathway alter learning and memory, and whether BDNF may mediate these events. Wild-type (WT) and BDNF heterozygous (+/-) mice were ovariectomized (OVX) at 5 weeks of age and simultaneously received empty, estradiol- or progesterone-filled implants for 7 weeks. At young adulthood, mice were tested for spatial and recognition memory in the Y-maze and novel-object recognition test, respectively. Hippocampal protein expression of BDNF and GABAergic interneuron markers, including parvalbumin, were assessed. WT OVX mice show impaired performance on Y-maze and novel-object recognition test. Estradiol replacement in OVX mice prevented the Y-maze impairment, a Behavioral abnormality of dorsal hippocampal origin. BDNF and parvalbumin protein expression in the dorsal hippocampus and parvalbumin-positive cell number in the dorsal CA1 were significantly reduced by OVX in WT mice, while E2 replacement prevented these deficits. In contrast, BDNF(+/-) mice showed either no response or an opposite response to hormone manipulation in both behavioral and molecular indices. Our data suggest that BDNF status is an important biomarker for predicting responsiveness to estrogenic compounds which have emerged as promising adjunctive therapeutics for schizophrenia patients.
Collapse
Affiliation(s)
- Y W C Wu
- Behavioural Neuroscience Laboratory, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, VIC, Australia
| | - X Du
- Behavioural Neuroscience Laboratory, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia
| | - M van den Buuse
- Behavioural Neuroscience Laboratory, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, VIC, Australia,School of Psychological Science, La Trobe University, Melbourne, VIC, Australia
| | - R A Hill
- Behavioural Neuroscience Laboratory, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia,Behavioural Neuroscience Laboratory, Florey Institute of Neuroscience and Mental Health, Kenneth Myer Building, Genetics Lane, Royal Parade, University of Melbourne, Parkville, VIC 3010, Australia. E-mail:
| |
Collapse
|
139
|
Gillies G, Virdee K, McArthur S, Dalley J. Sex-dependent diversity in ventral tegmental dopaminergic neurons and developmental programing: A molecular, cellular and behavioral analysis. Neuroscience 2014; 282:69-85. [PMID: 24943715 PMCID: PMC4245713 DOI: 10.1016/j.neuroscience.2014.05.033] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 05/12/2014] [Accepted: 05/18/2014] [Indexed: 02/02/2023]
Abstract
The knowledge that diverse populations of dopaminergic neurons within the ventral tegmental area (VTA) can be distinguished in terms of their molecular, electrophysiological and functional properties, as well as their differential projections to cortical and subcortical regions has significance for key brain functions, such as the regulation of motivation, working memory and sensorimotor control. Almost without exception, this understanding has evolved from landmark studies performed in the male sex. However, converging evidence from both clinical and pre-clinical studies illustrates that the structure and functioning of the VTA dopaminergic systems are intrinsically different in males and females. This may be driven by sex differences in the hormonal environment during adulthood ('activational' effects) and development (perinatal and/or pubertal 'organizational' effects), as well as genetic factors, especially the SRY gene on the Y chromosome in males, which is expressed in a sub-population of adult midbrain dopaminergic neurons. Stress and stress hormones, especially glucocorticoids, are important factors which interact with the VTA dopaminergic systems in order to achieve behavioral adaptation and enable the individual to cope with environmental change. Here, also, there is male/female diversity not only during adulthood, but also in early life when neurobiological programing by stress or glucocorticoid exposure differentially impacts dopaminergic developmental trajectories in male and female brains. This may have enduring consequences for individual resilience or susceptibility to pathophysiological change induced by stressors in later life, with potential translational significance for sex bias commonly found in disorders involving dysfunction of the mesocorticolimbic dopaminergic systems. These findings highlight the urgent need for a better understanding of the sexual dimorphism in the VTA if we are to improve strategies for the prevention and treatment of debilitating conditions which differentially affect men and women in their prevalence and nature, including schizophrenia, attention/deficit hyperactivity disorder, autism spectrum disorders, anxiety, depression and addiction.
Collapse
Affiliation(s)
- G.E. Gillies
- Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK,Corresponding author. Address: Division of Brain Sciences, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK. Tel: +44-(0)-20-7594-7050.
| | - K. Virdee
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge CB2 3EB, UK,Department of Psychology, University of Cambridge, Downing Street, Cambridge CB2 3EB, UK
| | - S. McArthur
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Charterhouse Square, London EC1 6BQ, UK
| | - J.W. Dalley
- Behavioural and Clinical Neuroscience Institute, University of Cambridge, Downing Street, Cambridge CB2 3EB, UK,Department of Psychology, University of Cambridge, Downing Street, Cambridge CB2 3EB, UK,Department of Psychiatry, University of Cambridge, Addenbrooke’s Hospital, Hill’s Road, Cambridge CB2 2QQ, UK
| |
Collapse
|
140
|
Kumar D, Thakur MK. Perinatal exposure to bisphenol-A impairs spatial memory through upregulation of neurexin1 and neuroligin3 expression in male mouse brain. PLoS One 2014; 9:e110482. [PMID: 25330104 PMCID: PMC4201550 DOI: 10.1371/journal.pone.0110482] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 09/21/2014] [Indexed: 11/19/2022] Open
Abstract
Bisphenol-A (BPA), a well known endocrine disruptor, impairs learning and memory in rodents. However, the underlying molecular mechanism of BPA induced impairment in learning and memory is not well known. As synaptic plasticity is the cellular basis of memory, the present study investigated the effect of perinatal exposure to BPA on the expression of synaptic proteins neurexin1 (Nrxn1) and neuroligin3 (Nlgn3), dendritic spine density and spatial memory in postnatal male mice. The pregnant mice were orally administered BPA (50 µg/kgbw/d) from gestation day (GD) 7 to postnatal day (PND) 21 and sesame oil was used as a vehicle control. In Morris water maze (MWM) test, BPA extended the escape latency time to locate the hidden platform in 8 weeks male mice. RT-PCR and Immunoblotting results showed significant upregulation of Nrxn1 and Nlgn3 expression in both cerebral cortex and hippocampus of 3 and 8 weeks male mice. This was further substantiated by in-situ hybridization and immunofluorescence techniques. BPA also significantly increased the density of dendritic spines in both regions, as analyzed by rapid Golgi staining. Thus our data suggest that perinatal exposure to BPA impairs spatial memory through upregulation of expression of synaptic proteins Nrxn1 and Nlgn3 and increased dendritic spine density in cerebral cortex and hippocampus of postnatal male mice.
Collapse
Affiliation(s)
- Dhiraj Kumar
- Biochemistry and Molecular Biology Laboratory, Brain Research Centre, Department of Zoology, Banaras Hindu University, Varanasi, India
| | - Mahendra Kumar Thakur
- Biochemistry and Molecular Biology Laboratory, Brain Research Centre, Department of Zoology, Banaras Hindu University, Varanasi, India
- * E-mail:
| |
Collapse
|
141
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: nuclear hormone receptors. Br J Pharmacol 2014; 170:1652-75. [PMID: 24528240 PMCID: PMC3892290 DOI: 10.1111/bph.12448] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. Nuclear hormone receptors are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, catalytic receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen P H Alexander
- School of Life Sciences, University of Nottingham Medical School, Nottingham, NG7 2UH, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Kireev RA, Vara E, Viña J, Tresguerres JAF. Melatonin and oestrogen treatments were able to improve neuroinflammation and apoptotic processes in dentate gyrus of old ovariectomized female rats. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9707. [PMID: 25135305 PMCID: PMC4453938 DOI: 10.1007/s11357-014-9707-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 08/11/2014] [Indexed: 06/03/2023]
Abstract
The aim of this study was to determine the outcomes of oestrogen and melatonin treatments following long-term ovarian hormone depletion on neuroinflammation and apoptotic processes in dentate gyrus of hippocampi. Forty-six female Wistar rats of 22 months of age were used. Twelve of them remained intact, and the other 34 were ovariectomized at 12 months of age. Ovariectomized animals were divided into three groups and treated for 10 weeks with oestrogens, melatonin or saline. All rats were killed by decapitation at 24 months of age, and dentate gyri were collected. A group of 2 month-old intact female rats was used as young control. The levels of pro-inflammatory cytokines and heat shock protein 70 (HSP 70) were analysed by ELISA. The expressions of TNFα, IL1β, GFAP, nNOS, iNOS, HO-1, NFκB, Bax, Bad, AIF, Bcl2 and SIRT1 genes were detected by real-time (RT)-PCR. Western blots were used to measure the protein expression of NFκB p65, NFκB p50/105, IκBα, IκBβ, p38 MAPK, MAP-2 and synapsin I. We have assessed the ability of 17β-oestradiol and melatonin administration to downregulate markers of neuroinflammation in the dentate gyrus of ovariectomized female rats. Results indicated that 17β-oestradiol and melatonin treatments were able to significantly decrease expression of pro-inflammatory cytokines, iNOS and HO-1 in the hippocampus when compared to non-treated animals. A similar age- and long-term ovarian hormone depletion- related increase in GFAP was also attenuated after both melatonin and oestradiol treatments. In a similar way to oestradiol, melatonin decreased the activation of p38 MAPK and NFκB pathways. The treatments enhanced the levels of synaptic molecules synapsin I and MAP-2 and have been shown to modulate the pro-antiapoptotic ratio favouring the second and to increase SIRT1 expression. These findings support the potential therapeutic role of melatonin and oestradiol as protective anti-inflammatory agents for the central nervous system during menopause.
Collapse
Affiliation(s)
- Roman A Kireev
- Instituto de Investigación Biomédica de Vigo (IBIV), Xerencia de Xestión Integrada de Vigo, SERGAS, Biomedical Research Unit, Hospital Rebullón (CHUVI), Puxeiros s/n, 36415, MOS Pontevedra, Spain,
| | | | | | | |
Collapse
|
143
|
Conroy SK, McDonald BC, Ahles TA, West JD, Saykin AJ. Chemotherapy-induced amenorrhea: a prospective study of brain activation changes and neurocognitive correlates. Brain Imaging Behav 2014; 7:491-500. [PMID: 23793983 DOI: 10.1007/s11682-013-9240-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Chemotherapy-induced amenorrhea (CIA) often occurs in pre- and peri-menopausal BC patients, and while cancer/chemotherapy and abrupt estrogen loss have separately been shown to affect cognition and brain function, studies of the cognitive effects of CIA are equivocal, and its effects on brain function are unknown. Functional MRI (fMRI) during a working memory task was used to prospectively assess the pattern of brain activation and deactivation prior to and 1 month after chemotherapy in BC patients who experienced CIA (n = 9), post-menopausal BC patients undergoing chemotherapy (n = 9), and pre- and post-menopausal healthy controls (n = 6 each). Neurocognitive testing was also performed at both time points. Repeated measures general linear models were used to assess statistical significance, and age was a covariate in all analyses. We observed a group-by-time interaction in the combined magnitudes of brain activation and deactivation (p = 0.006): the CIA group increased in magnitude from baseline to post-treatment while other groups maintained similar levels over time. Further, the change in brain activity magnitude in CIA was strongly correlated with change in processing speed neurocognitive testing score (r = 0.837 p = 0.005), suggesting this increase in brain activity reflects effective cognitive compensation. Our results demonstrate prospectively that the pattern of change in brain activity from pre- to post-chemotherapy varies according to pre-treatment menopausal status. Cognitive correlates add to the potential clinical significance of these findings. These findings have implications for risk appraisal and development of prevention or treatment strategies for cognitive changes in CIA.
Collapse
Affiliation(s)
- Susan K Conroy
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, 355 W. 16th St., GH Suite 4100, Indianapolis, IN, 46202, USA
| | | | | | | | | |
Collapse
|
144
|
Periodic Estrogen Receptor-Beta Activation: A Novel Approach to Prevent Ischemic Brain Damage. Neurochem Res 2014; 40:2009-17. [PMID: 24906488 DOI: 10.1007/s11064-014-1346-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 05/12/2014] [Accepted: 05/22/2014] [Indexed: 02/06/2023]
Abstract
In women, the risk for cerebral ischemia climbs rapidly after menopause. At menopause, production of ovarian hormones; i.e., progesterone and estrogen, slowly diminishes. Estrogen has been suggested to confer natural protection to premenopausal women from ischemic stroke and some of its debilitating consequences. This notion is also strongly supported by laboratory studies showing that a continuous chronic 17β-estradiol (E2; a potent estrogen) regimen protects brain from ischemic injury. However, concerns regarding the safety of the continuous intake of E2 were raised by the failed translation to the clinic. Recent studies demonstrated that repetitive periodic E2 pretreatments, in contrast to continuous E2 treatment, provided neuroprotection against cerebral ischemia in ovariectomized rats. Periodic E2 pretreatment protects hippocampal neurons through activation of estrogen receptor subtype beta (ER-β). Apart from neuroprotection, periodic activation of ER-β in ovariectomized rats significantly improves hippocampus-dependent learning and memory. Difficulties in learning and memory loss are the major consequence of ischemic brain damage. Periodic ER-β agonist pretreatment may provide pharmacological access to a protective state against ischemic stroke and its debilitating consequences. The use of ER-β-selective agonists constitutes a safer target for future research than ER-α agonist or E2, inasmuch as it lacks the ability to stimulate the proliferation of breast or endometrial tissue. In this review, we highlight ER-β signaling as a guide for future translational research to reduce cognitive decline and cerebral ischemia incidents/impact in post-menopausal women, while avoiding the side effects produced by chronic E2 treatment.
Collapse
|
145
|
Pratchayasakul W, Chattipakorn N, Chattipakorn SC. Estrogen restores brain insulin sensitivity in ovariectomized non-obese rats, but not in ovariectomized obese rats. Metabolism 2014; 63:851-9. [PMID: 24742706 DOI: 10.1016/j.metabol.2014.03.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 02/26/2014] [Accepted: 03/17/2014] [Indexed: 11/21/2022]
Abstract
OBJECTIVE We previously demonstrated that obesity caused the reduction of peripheral and brain insulin sensitivity and that estrogen therapy improved these defects. However, the beneficial effect of estrogen on brain insulin sensitivity and oxidative stress in either ovariectomy alone or ovariectomy with obesity models has not been determined. We hypothesized that ovariectomy alone or ovariectomy with obesity reduces brain insulin sensitivity and increases brain oxidative stress, which are reversed by estrogen treatment. MATERIALS/METHODS Thirty female rats were assigned as either sham-operated or ovariectomized. After the surgery, each group was fed either a normal diet or high-fat diet for 12 weeks. At week 13, rats in each group received either the vehicle or estradiol for 30 days. At week 16, blood and brain were collected for determining the peripheral and brain insulin sensitivity as well as brain oxidative stress. RESULTS We found that ovariectomized rats and high-fat diet fed rats incurred obesity, reduced peripheral and brain insulin sensitivity, and increased brain oxidative stress. Estrogen ameliorated peripheral insulin sensitivity in these rats. However, the beneficial effect of estrogen on brain insulin sensitivity and brain oxidative stress was observed only in ovariectomized normal diet-fed rats, but not in ovariectomized high fat diet-fed rats. CONCLUSIONS Our results suggested that reduced brain insulin sensitivity and increased brain oxidative stress occurred after either ovariectomy or obesity. However, the reduced brain insulin sensitivity and the increased brain oxidative stress in ovariectomy with obesity could not be ameliorated by estrogen treatment.
Collapse
Affiliation(s)
- Wasana Pratchayasakul
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Department of Oral Biology and Diagnostic Science, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
146
|
Li R, Cui J, Shen Y. Brain sex matters: estrogen in cognition and Alzheimer's disease. Mol Cell Endocrinol 2014; 389:13-21. [PMID: 24418360 PMCID: PMC4040318 DOI: 10.1016/j.mce.2013.12.018] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 12/31/2013] [Accepted: 12/31/2013] [Indexed: 02/06/2023]
Abstract
Estrogens are the primary female sex hormones and play important roles in both reproductive and non-reproductive systems. Estrogens can be synthesized in non-reproductive tissues such as liver, heart, muscle, bone and the brain. During the past decade, increasing evidence suggests that brain estrogen can not only be synthesized by neurons, but also by astrocytes. Brain estrogen also works locally at the site of synthesis in paracrine and/or intracrine fashion to maintain important tissue-specific functions. Here, we will focus on the biology of brain estrogen and its impact on cognitive function and Alzheimer's disease. This comprehensive review provides new insights into brain estrogens by presenting a better understanding of the tissue-specific estrogen effects and their roles in healthy ageing and cognitive function.
Collapse
Affiliation(s)
- Rena Li
- Center for Hormone Advanced Science and Education (CHASE), Roskamp Institute, Sarasota, FL 34243, United States.
| | - Jie Cui
- Center for Hormone Advanced Science and Education (CHASE), Roskamp Institute, Sarasota, FL 34243, United States
| | - Yong Shen
- Center for Advanced Therapeutic Strategies for Brain Disorders (CATSBD), Roskamp Institute, Sarasota, FL 34243, United States
| |
Collapse
|
147
|
Lynch G, Cox CD, Gall CM. Pharmacological enhancement of memory or cognition in normal subjects. Front Syst Neurosci 2014; 8:90. [PMID: 24904313 PMCID: PMC4033242 DOI: 10.3389/fnsys.2014.00090] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 04/30/2014] [Indexed: 12/14/2022] Open
Abstract
The possibility of expanding memory or cognitive capabilities above the levels in high functioning individuals is a topic of intense discussion among scientists and in society at large. The majority of animal studies use behavioral endpoint measures; this has produced valuable information but limited predictability for human outcomes. Accordingly, several groups are pursuing a complementary strategy with treatments targeting synaptic events associated with memory encoding or forebrain network operations. Transcription and translation figure prominently in substrate work directed at enhancement. Notably, the question of why new proteins would be needed for a now-forming memory given that learning-driven synthesis presumably occurred throughout the immediate past has been largely ignored. Despite this conceptual problem, and some controversy, recent studies have reinvigorated the idea that selective gene manipulation is a plausible route to enhancement. Efforts to improve memory by facilitating synaptic encoding of information have also progressed, in part due of breakthroughs on mechanisms that stabilize learning-related, long-term potentiation (LTP). These advances point to a reductionistic hypothesis for a diversity of experimental results on enhancement, and identify under-explored possibilities. Cognitive enhancement remains an elusive goal, in part due to the difficulty of defining the target. The popular view of cognition as a collection of definable computations seems to miss the fluid, integrative process experienced by high functioning individuals. The neurobiological approach obviates these psychological issues to directly test the consequences of improving throughput in networks underlying higher order behaviors. The few relevant studies testing drugs that selectively promote excitatory transmission indicate that it is possible to expand cortical networks engaged by complex tasks and that this is accompanied by capabilities not found in normal animals.
Collapse
Affiliation(s)
- Gary Lynch
- Department of Psychiatry and Human Behavior, University of California Irvine, CA, USA ; Department of Anatomy and Neurobiology, University of California Irvine, CA, USA
| | - Conor D Cox
- Department of Anatomy and Neurobiology, University of California Irvine, CA, USA
| | - Christine M Gall
- Department of Anatomy and Neurobiology, University of California Irvine, CA, USA
| |
Collapse
|
148
|
Lipatova O, Byrd D, Green JT, Toufexis DJ. Effects of continuous vs. cycling estrogen replacement on the acquisition, retention and expression of place- and response-learning in the open-field tower maze. Neurobiol Learn Mem 2014; 114:81-9. [PMID: 24837787 DOI: 10.1016/j.nlm.2014.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 04/23/2014] [Accepted: 05/05/2014] [Indexed: 01/28/2023]
Abstract
Estrogen has been shown to either enhance or impair learning and memory in female rats. The use of different experimental paradigms or estrogen treatment regimens may contribute to these disparate findings. In order to assess the effect of different estradiol (E2) treatments on several aspects of cognition, we trained ovariectomized female rats with either continuous, cycling, or vehicle E2 replacement, in an open-field tower maze task (OFTM) designed to test reference memory in a low-stress environment. In addition, in order to compare two distinct learning and memory systems, rats were trained to use either a dorsolateral striatum-based response type learning or a hippocampal-based place type learning to solve the maze. Results showed that cyclic, but not continuous, E2 replacement facilitated the acquisition of spatial memory in place-learners. Neither E2 regimen affected acquisition in response-learners. Additionally, when all experimental groups were performing at asymptote, rats were evaluated for performance stability by changing the location of their start position in the OFTM. Both regimens of E2 disrupted the expression of spatial memory in place-learners following the novel start position. However, E2 replacement protected ovariectomized female rats from the disruption of memory expression following a start position change in response-learners. Additionally all experimental groups performed equally well when tested following a 21-day period during which rats were absent from the maze. These results suggest that E2 fluctuation is particularly important in the acquisition of hippocampal-mediated spatial learning, and that hippocampal-based memory may be subject to disruption following environmental change, while striatum-based memory is subject to protection.
Collapse
Affiliation(s)
- Olga Lipatova
- University of Vermont, Department of Psychology, 2 Colchester Ave., Burlington, VT 05405, United States.
| | - Dennis Byrd
- University of Vermont, Department of Psychology, 2 Colchester Ave., Burlington, VT 05405, United States
| | - John T Green
- University of Vermont, Department of Psychology, 2 Colchester Ave., Burlington, VT 05405, United States
| | - Donna J Toufexis
- University of Vermont, Department of Psychology, 2 Colchester Ave., Burlington, VT 05405, United States
| |
Collapse
|
149
|
Zhao L, Mao Z, Chen S, Schneider LS, Brinton RD. Early intervention with an estrogen receptor β-selective phytoestrogenic formulation prolongs survival, improves spatial recognition memory, and slows progression of amyloid pathology in a female mouse model of Alzheimer's disease. J Alzheimers Dis 2014; 37:403-19. [PMID: 23948892 DOI: 10.3233/jad-122341] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Our recent developments have yielded a novel phytoestrogenic formulation, referred to as the phyto-β-SERM formulation, which exhibits an 83-fold binding selectivity for the estrogen receptor subtype β (ERβ) over ERα. Earlier studies indicate that the phyto-β-SERM formulation is neuroprotective and promotes estrogenic mechanisms in the brain while devoid of feminizing activity in the periphery. Further investigation in a mouse model of human menopause indicates that chronic exposure to the phyto-β-SERM formulation at a clinically relevant dosage prevents/alleviates menopause-related climacteric symptoms. This study assessed the efficacy, in an early intervention paradigm, of the phyto-β-SERM formulation in the regulation of early stages of physical and neurological changes associated with Alzheimer's disease (AD) in a female triple transgenic mouse model of AD. Results demonstrated that, when initiated prior to the appearance of AD pathology, a 9-month dietary supplementation with the phyto-β-SERM formulation promoted physical health, prolonged survival, improved spatial recognition memory, and attenuated amyloid-β deposition and plaque formation in the brains of treated AD mice. In comparison, dietary supplementation of a commercial soy extract preparation showed no effect on cognitive measures, although it appeared to have a positive impact on amyloid pathology. In overall agreement with the behavioral and histological outcomes, results from a gene expression profiling analysis offered insights on the underlying molecular mechanisms associated with the two dietary treatments. In particular, the data suggests that there may be a crosstalk between ERβ and glycogen synthase kinase 3 signaling pathways that could play a role in conferring ERβ-mediated neuroprotection against AD. Taken together, these results support the therapeutic potential of the phyto-β-SERM formulation for prevention and/or early intervention of AD, and warrants further investigations in human studies.
Collapse
Affiliation(s)
- Liqin Zhao
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | | | | | | | | |
Collapse
|
150
|
Irwin RW, Brinton RD. Allopregnanolone as regenerative therapeutic for Alzheimer's disease: Translational development and clinical promise. Prog Neurobiol 2014; 113:40-55. [PMID: 24044981 PMCID: PMC10124616 DOI: 10.1016/j.pneurobio.2013.08.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 08/22/2013] [Accepted: 08/25/2013] [Indexed: 10/26/2022]
Abstract
Herein, we review a translational development plan to advance allopregnanolone to the clinic as a regenerative therapeutic for neurodegenerative diseases, in particular Alzheimer's. Allopregnanolone, an endogenous neurosteroid that declines with age and neurodegenerative disease, was exogenously administered and assessed for safety and efficacy to promote neuro-regeneration, cognitive function and reduction of Alzheimer's pathology. Allopregnanolone-induced neurogenesis correlated with restoration of learning and memory function in a mouse model of Alzheimer's disease and was comparably efficacious in aged normal mice. Critical to success was a dosing and treatment regimen that was consistent with the temporal requirements of systems biology of regeneration in brain. A treatment regimen that adhered to regenerative requirements of brain was also efficacious in reducing Alzheimer's pathology. With an optimized dosing and treatment regimen, chronic allopregnanolone administration promoted neurogenesis, oligodendrogenesis, reduced neuroinflammation and beta-amyloid burden while increasing markers of white matter generation and cholesterol homeostasis. Allopregnanolone meets three of the four drug-like physicochemical properties described by Lipinski's rule that predict the success rate of drugs in development for clinical trials. Pharmacokinetic and pharmacodynamic outcomes, securing GMP material, development of clinically translatable formulations and acquiring regulatory approval are discussed. Investigation of allopregnanolone as a regenerative therapeutic has provided key insights into mechanistic targets for neurogenesis and disease modification, dosing requirements, optimal treatment regimen, route of administration and the appropriate formulation necessary to advance to proof of concept clinical studies to determine efficacy of allopregnanolone as a regenerative and disease modifying therapeutic for Alzheimer's disease.
Collapse
|